1
|
Mahmud J, Geiler BW, Biswas J, Miller MJ, Myers JE, Matthews SM, Wass AB, O’Connor CM, Chan GC. Delivery of US28 by incoming HCMV particles rapidly attenuates Akt activity to suppress HCMV lytic replication in monocytes. Sci Signal 2024; 17:eadn8727. [PMID: 39190708 PMCID: PMC11460310 DOI: 10.1126/scisignal.adn8727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024]
Abstract
Establishing a nonproductive, quiescent infection within monocytes is essential for the spread of human cytomegalovirus (HCMV). We investigated the mechanisms through which HCMV establishes a quiescent infection in monocytes. US28 is a virally encoded G protein-coupled receptor (GPCR) that is essential for silent infections within cells of the myeloid lineage. We found that preformed US28 was rapidly delivered to monocytes by HCMV viral particles, whereas the de novo synthesis of US28 was delayed for several days. A recombinant mutant virus lacking US28 (US28Δ) was unable to establish a quiescent infection, resulting in a fully productive lytic infection able to produce progeny virus. Infection with US28Δ HCMV resulted in the phosphorylation of the serine and threonine kinase Akt at Ser473 and Thr308, in contrast with the phosphorylation of Akt only at Ser473 after WT viral infection. Inhibiting the dual phosphorylation of Akt prevented the lytic replication of US28Δ, and ectopic expression of a constitutively phosphorylated Akt variant triggered lytic replication of wild-type HCMV. Mechanistically, we found that US28 was necessary and sufficient to attenuate epidermal growth factor receptor (EGFR) signaling induced during the entry of WT virus, which led to the site-specific phosphorylation of Akt at Ser473. Thus, particle-delivered US28 fine-tunes Akt activity by limiting HCMV-induced EGFR activation during viral entry, enabling quiescent infection in monocytes.
Collapse
Affiliation(s)
- Jamil Mahmud
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Brittany W. Geiler
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Juthi Biswas
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Michael J. Miller
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Julia E. Myers
- Infection Biology, Lerner Research Institute, Sheikha Fatima bint Mubarak Global Center for Pathogen & Human Health Research, Cleveland Clinic, Cleveland, OH 44195
- Case Comprehensive Cancer Center, Cleveland, OH 44106
| | - Stephen M. Matthews
- Infection Biology, Lerner Research Institute, Sheikha Fatima bint Mubarak Global Center for Pathogen & Human Health Research, Cleveland Clinic, Cleveland, OH 44195
- Case Comprehensive Cancer Center, Cleveland, OH 44106
| | - Amanda B. Wass
- Infection Biology, Lerner Research Institute, Sheikha Fatima bint Mubarak Global Center for Pathogen & Human Health Research, Cleveland Clinic, Cleveland, OH 44195
- Case Comprehensive Cancer Center, Cleveland, OH 44106
| | - Christine M. O’Connor
- Infection Biology, Lerner Research Institute, Sheikha Fatima bint Mubarak Global Center for Pathogen & Human Health Research, Cleveland Clinic, Cleveland, OH 44195
- Case Comprehensive Cancer Center, Cleveland, OH 44106
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195
| | - Gary C. Chan
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
2
|
Zeisbrich M, Rzepka R, Finzel S, Venhoff N, Voll RE. Macrophage colony-stimulating factor receptor/CD115 + non-classical monocytes are expanded in systemic lupus erythematosus and associated with lupus nephritis. Scand J Rheumatol 2024:1-10. [PMID: 39171822 DOI: 10.1080/03009742.2024.2387483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
OBJECTIVE In systemic lupus erythematosus (SLE), the non-classical monocyte compartment is expanded, but its phenotype and association with clinical disease manifestations have not been explored. METHOD Monocyte subsets from 39 SLE patients, 32 healthy age-matched controls, and 16 patients from a disease control (autoimmune connective tissue disease other than SLE) were determined based on CD14 and CD16 surface expression. Cell surface expression of the receptors for macrophage colony-stimulating factor (M-CSF) (CD115) and granulocyte-macrophage colony-stimulating factor (GM-CSF) (CD116), as well as 6-Sulpho LacNAc (slan), were analysed by flow cytometry. The association of monocyte populations with disease manifestations, disease activity markers, and current medication of each patient was analysed by chart review. RESULTS Non-classical monocytes displayed a cell-type specific signature of high M-CSF receptor CD115 and low GM-CSF receptor CD116 expression that separated them from the other two monocyte subsets. In healthy individuals, the M-CSF receptor on non-classical monocytes was an age-dependent surface marker, with lower expression in young adults. However, SLE monocytes were characterized by a marked expansion of M-CSF receptor/CD115+ non-classical monocytes in patients of all ages. The expanded population of M-CSF receptor/CD115+ non-classical monocytes was associated with lupus nephritis but not with disease activity, and coexpressed slan. CONCLUSION The non-classical monocyte subset in SLE is characterized by an expansion of M-CSF receptor/CD115+ cells that are associated with lupus nephritis and coexpress slan.
Collapse
Affiliation(s)
- M Zeisbrich
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - R Rzepka
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - S Finzel
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - N Venhoff
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - R E Voll
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
3
|
Yurakova TR, Gorshkova EA, Nosenko MA, Drutskaya MS. Metabolic Adaptations and Functional Activity of Macrophages in Homeostasis and Inflammation. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:817-838. [PMID: 38880644 DOI: 10.1134/s0006297924050043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 06/18/2024]
Abstract
In recent years, the role of cellular metabolism in immunity has come into the focus of many studies. These processes form a basis for the maintenance of tissue integrity and homeostasis, as well as represent an integral part of the immune response, in particular, inflammation. Metabolic adaptations not only ensure energy supply for immune response, but also affect the functions of immune cells by controlling transcriptional and post-transcriptional programs. Studying the immune cell metabolism facilitates the search for new treatment approaches, especially for metabolic disorders. Macrophages, innate immune cells, are characterized by a high functional plasticity and play a key role in homeostasis and inflammation. Depending on the phenotype and origin, they can either perform various regulatory functions or promote inflammation state, thus exacerbating the pathological condition. Furthermore, their adaptations to the tissue-specific microenvironment influence the intensity and type of immune response. The review examines the effect of metabolic reprogramming in macrophages on the functional activity of these cells and their polarization. The role of immunometabolic adaptations of myeloid cells in tissue homeostasis and in various pathological processes in the context of inflammatory and metabolic diseases is specifically discussed. Finally, modulation of the macrophage metabolism-related mechanisms reviewed as a potential therapeutic approach.
Collapse
Affiliation(s)
- Taisiya R Yurakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Ekaterina A Gorshkova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Maxim A Nosenko
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, D02F306, Ireland
| | - Marina S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia
| |
Collapse
|
4
|
Mahmud J, Geiler BW, Biswas J, Miller MJ, Myers JE, Matthews SM, Wass AB, O'Connor CM, Chan GC. Virion-associated US28 rapidly modulates Akt activity to suppress HCMV lytic replication in monocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.05.556359. [PMID: 37732204 PMCID: PMC10508783 DOI: 10.1101/2023.09.05.556359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Establishing a non-productive quiescent/silent infection within monocytes is essential for spread of human cytomegalovirus (HCMV). Yet, how HCMV establishes a quiescent infection in monocytes remains unclear. US28 is a viral G protein-coupled receptor (GPCR) essential for silent infections within cells of the myeloid lineage. We found virion-associated US28 was rapidly delivered to monocytes, while de novo synthesized US28 was delayed for several days. A recombinant mutant virus lacking US28 (US28Δ) was unable to establish a quiescent infection, resulting in a fully productive lytic replication cycle. Mechanistically, viral entry of US28Δ phosphorylated Akt at both serine 473 (S473) and threonine 308 (T308), which contrasted with the site-specific phosphorylation of Akt at S473 following WT infection. Preventing Akt bi-phosphorylation prevented lytic replication of US28Δ, and ectopic expression of a constitutively phosphorylated Akt variant triggered lytic replication of WT infection. Our data demonstrate that virion-delivered US28 fine-tunes Akt activity to permit HCMV infection to enter a quiescent state following primary infection of monocytes.
Collapse
|
5
|
Kapanadze T, Gamrekelashvili J, Sablotny S, Kijas D, Haller H, Schmidt-Ott K, Limbourg FP. CSF-1 and Notch signaling cooperate in macrophage instruction and tissue repair during peripheral limb ischemia. Front Immunol 2023; 14:1240327. [PMID: 37691936 PMCID: PMC10484478 DOI: 10.3389/fimmu.2023.1240327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Ischemia causes an inflammatory response featuring monocyte-derived macrophages (MF) involved in angiogenesis and tissue repair. Angiogenesis and ischemic macrophage differentiation are regulated by Notch signaling via Notch ligand Delta-like 1 (Dll1). Colony stimulating factor 1 (CSF-1) is an essential MF lineage factor, but its role in ischemic macrophage development and the interaction with Notch signaling is so far unclear. Using a mouse model of hind limb ischemia with CSF-1 inhibitor studies and Dll1 heterozygous mice we show that CSF-1 is induced in the ischemic niche by a subpopulation of stromal cells expressing podoplanin, which was paralleled by the development of ischemic macrophages. Inhibition of CSF-1 signaling with small molecules or blocking antibodies impaired macrophage differentiation but prolonged the inflammatory response, resulting in impaired perfusion recovery and tissue regeneration. Yet, despite high levels of CSF-1, macrophage maturation and perfusion recovery were impaired in mice with Dll1 haploinsufficiency, while inflammation was exaggerated. In vitro, CSF-1 was not sufficient to induce full MF differentiation from donor monocytes in the absence of recombinant DLL1, while the presence of DLL1 in a dose-dependent manner stimulated MF differentiation in combination with CSF-1. Thus, CSF-1 is an ischemic niche factor that cooperates with Notch signaling in a non-redundant fashion to instruct macrophage cell fate and maturation, which is required for ischemic perfusion recovery and tissue repair.
Collapse
Affiliation(s)
- Tamar Kapanadze
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Jaba Gamrekelashvili
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Stefan Sablotny
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Dustin Kijas
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Kai Schmidt-Ott
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Florian P. Limbourg
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| |
Collapse
|
6
|
Fu XZ, Wang Y. Interferon-γ regulates immunosuppression in septic mice by promoting the Warburg effect through the PI3K/AKT/mTOR pathway. Mol Med 2023; 29:95. [PMID: 37434129 PMCID: PMC10337057 DOI: 10.1186/s10020-023-00690-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 06/20/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND The main cause of high mortality from sepsis is that immunosuppression leads to life-threatening organ dysfunction, and reversing immunosuppression is key to sepsis treatment. Interferon γ (IFNγ) is a potential therapy for immunosuppression of sepsis, promoting glycolysis to restore metabolic defects in monocytes, but the mechanism of treatment is unclear. METHODS To explore the immunotherapeutic mechanism of IFNγ, this study linked the Warburg effect (aerobic glycolysis) to immunotherapy for sepsis and used cecal ligation perforation (CLP) and lipopolysaccharide (LPS) to stimulate dendritic cells (DC) to establish in vivo and in vitro sepsis models, Warburg effect inhibitors (2-DG) and PI3K pathway inhibitors (LY294002) were used to explore the mechanism by which IFNγ regulates immunosuppression in mice with sepsis through the Warburg effect. RESULTS IFNγ markedly inhibited the reduction in cytokine secretion from lipopolysaccharide (LPS)-stimulated splenocytes. IFNγ-treated mice had significantly increased the percentages of positive costimulatory receptor CD86 on Dendritic cells expressing and expression of splenic HLA-DR. IFNγ markedly reduced DC-cell apoptosis by upregulating the expression of Bcl-2 and downregulating the expression of Bax. CLP-induced formation of regulatory T cells in the spleen was abolished in IFNγ -treated mice. IFNγ treatment reduced the expression of autophagosomes in DC cells. IFNγ significant reduce the expression of Warburg effector-related proteins PDH, LDH, Glut1, and Glut4, and promote glucose consumption, lactic acid, and intracellular ATP production. After the use of 2-DG to suppress the Warburg effect, the therapeutic effect of IFNγ was suppressed, demonstrating that IFNγ reverses immunosuppression by promoting the Warburg effect. Moreover, IFNγ increased the expression of phosphoinositide 3-kinases (PI3K), protein kinase B (Akt), rapamycin target protein (mTOR), hypoxia-inducible factor-1 (HIF-1α), pyruvate dehydrogenase kinase (PDK1) protein, the use of 2-DG and LY294002 can inhibit the expression of the above proteins, LY294002 also inhibits the therapeutic effect of IFNγ. CONCLUSIONS It was finally proved that IFNγ promoted the Warburg effect through the PI3K/Akt/mTOR pathway to reverse the immunosuppression caused by sepsis. This study elucidates the potential mechanism of the immunotherapeutic effect of IFNγ in sepsis, providing a new target for the treatment of sepsis.
Collapse
Affiliation(s)
- Xu-Zhe Fu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
7
|
Curson JE, Liu L, Luo L, Muusse TW, Lucas RM, Gunther KS, Vajjhala PR, Abrol R, Jones A, Kapetanovic R, Stacey KJ, Stow JL, Sweet MJ. TLR4 phosphorylation at tyrosine 672 activates the ERK/c-FOS signaling module for LPS-induced cytokine responses in macrophages. Eur J Immunol 2023; 53:e2250056. [PMID: 37058370 PMCID: PMC10947571 DOI: 10.1002/eji.202250056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 03/20/2023] [Accepted: 04/11/2023] [Indexed: 04/15/2023]
Abstract
TLRs engage numerous adaptor proteins and signaling molecules, enabling a complex series of post-translational modifications (PTMs) to mount inflammatory responses. TLRs themselves are post-translationally modified following ligand-induced activation, with this being required to relay the full spectrum of proinflammatory signaling responses. Here, we reveal indispensable roles for TLR4 Y672 and Y749 phosphorylation in mounting optimal LPS-inducible inflammatory responses in primary mouse macrophages. LPS promotes phosphorylation at both tyrosine residues, with Y749 phosphorylation being required for maintenance of total TLR4 protein levels and Y672 phosphorylation exerting its pro-inflammatory effects more selectively by initiating ERK1/2 and c-FOS phosphorylation. Our data also support a role for the TLR4-interacting membrane proteins SCIMP and the SYK kinase axis in mediating TLR4 Y672 phosphorylation to permit downstream inflammatory responses in murine macrophages. The corresponding residue in human TLR4 (Y674) is also required for optimal LPS signaling responses. Our study, thus, reveals how a single PTM on one of the most widely studied innate immune receptors orchestrates downstream inflammatory responses.
Collapse
Affiliation(s)
- James E.B. Curson
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Liping Liu
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Lin Luo
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Timothy W. Muusse
- School of Chemistry and Molecular Biosciences (SCMB) and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Richard M. Lucas
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Kimberley S. Gunther
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Parimala R. Vajjhala
- School of Chemistry and Molecular Biosciences (SCMB) and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Rishika Abrol
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Alun Jones
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Katryn J. Stacey
- School of Chemistry and Molecular Biosciences (SCMB) and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Jennifer L. Stow
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Matthew J. Sweet
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
8
|
Bahlmann LC, Xue C, Chin AA, Skirzynska A, Lu J, Thériault B, Uehling D, Yerofeyeva Y, Peters R, Liu K, Chen J, Martel AL, Yaffe M, Al-Awar R, Goswami RS, Ylanko J, Andrews DW, Kuruvilla J, Laister RC, Shoichet MS. Targeting tumour-associated macrophages in hodgkin lymphoma using engineered extracellular matrix-mimicking cryogels. Biomaterials 2023; 297:122121. [PMID: 37075613 DOI: 10.1016/j.biomaterials.2023.122121] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/08/2023] [Accepted: 04/06/2023] [Indexed: 04/21/2023]
Abstract
Tumour-associated macrophages are linked with poor prognosis and resistance to therapy in Hodgkin lymphoma; however, there are no suitable preclinical models to identify macrophage-targeting therapeutics. We used primary human tumours to guide the development of a mimetic cryogel, wherein Hodgkin (but not Non-Hodgkin) lymphoma cells promoted primary human macrophage invasion. In an invasion inhibitor screen, we identified five drug hits that significantly reduced tumour-associated macrophage invasion: marimastat, batimastat, AS1517499, ruxolitinib, and PD-169316. Importantly, ruxolitinib has demonstrated recent success in Hodgkin lymphoma clinical trials. Both ruxolitinib and PD-169316 (a p38 mitogen-activated protein kinase (p38 MAPK) inhibitor) decreased the percent of M2-like macrophages; however, only PD-169316 enhanced the percentage of M1-like macrophages. We validated p38 MAPK as an anti-invasion drug target with five additional drugs using a high-content imaging platform. With our biomimetic cryogel, we modeled macrophage invasion in Hodgkin lymphoma and then used it for target discovery and drug screening, ultimately identifying potential future therapeutics.
Collapse
Affiliation(s)
- Laura C Bahlmann
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
| | - Chang Xue
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
| | - Allysia A Chin
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
| | - Arianna Skirzynska
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
| | - Joy Lu
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
| | - Brigitte Thériault
- Drug Discovery Program, Ontario Institute of Cancer Research, 661 University Ave Suite 510, Toronto, Ontario, M5G 0A3, Canada
| | - David Uehling
- Drug Discovery Program, Ontario Institute of Cancer Research, 661 University Ave Suite 510, Toronto, Ontario, M5G 0A3, Canada
| | - Yulia Yerofeyeva
- Biomarker Imaging Research Laboratory, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Rachel Peters
- Biomarker Imaging Research Laboratory, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Kela Liu
- Biomarker Imaging Research Laboratory, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Jianan Chen
- Department of Medical Biophysics, University of Toronto, 101 College St Suite 15-701, Toronto, Ontario, M5G 1L7, Canada
| | - Anne L Martel
- Department of Medical Biophysics, University of Toronto, 101 College St Suite 15-701, Toronto, Ontario, M5G 1L7, Canada; Physical Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Martin Yaffe
- Department of Medical Biophysics, University of Toronto, 101 College St Suite 15-701, Toronto, Ontario, M5G 1L7, Canada; Physical Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Rima Al-Awar
- Drug Discovery Program, Ontario Institute of Cancer Research, 661 University Ave Suite 510, Toronto, Ontario, M5G 0A3, Canada; Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Rashmi S Goswami
- Biological Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada; Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Jarkko Ylanko
- Biological Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - David W Andrews
- Department of Medical Biophysics, University of Toronto, 101 College St Suite 15-701, Toronto, Ontario, M5G 1L7, Canada; Biological Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada; Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - John Kuruvilla
- Princess Margaret Cancer Centre, University Health Network, 610 University Ave, Toronto, Ontario, M5G 2C1, Canada
| | - Rob C Laister
- Princess Margaret Cancer Centre, University Health Network, 610 University Ave, Toronto, Ontario, M5G 2C1, Canada.
| | - Molly S Shoichet
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada.
| |
Collapse
|
9
|
Gallo CC, Honda TSB, Alves PT, Han SW. Macrophages mobilized by the overexpression of the macrophage-colony stimulating factor promote efficient recovery of the ischemic muscle functionality. Life Sci 2023; 317:121478. [PMID: 36758666 DOI: 10.1016/j.lfs.2023.121478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/24/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
AIMS Narrowing or occlusion of arteries that supply the limbs can evolve to critical limb ischemia. M-CSF promotes proliferation, differentiation and survival of monocytes and macrophages, and polarization of macrophages to M2-subtype, which are essential elements for vessel formation and tissue repair. Based on these properties of M-CSF, we hypothesize that transfection of M-CSF into ischemic limbs may promote vessel formation and repair of ischemic limbs. MAIN METHODS Hindlimb ischemia was surgically induced in 10-12 weeks old Balb/c and gene therapy was performed with intramuscular application of either uP-MCSF or uP plasmids (100 μg). Macrophage and monocyte subpopulations were assessed by flow cytometry and blood flow was monitored by Laser Doppler Perfusion Imaging (LDPI). Thirty days after transfection, we assessed gastrocnemius mass and muscle force, subsequently collecting the muscle for histology. KEY FINDINGS We successfully developed the uP-MCSF plasmid, which increases M-CSF expression in the muscle transiently. Thirty days after uP-MCSF gene therapy in ischemic muscles, the treated group presented: improved muscle force, reduced fibrosis and increased arteriogenesis, although LDPI analysis did not show any significant difference in blood flow among groups. Noteworthy, we observed a temporary increase in MHCIIhighCD206high macrophages after uP-MCSF transfection. SIGNIFICANCE M-CSF gene therapy improved ischemic muscle functionality by promoting arteriogenesis and decreasing fibrosis, likely through increased MHCIIhighCD206high macrophages and not via classically known M2-macrophages.
Collapse
Affiliation(s)
- Camila Congentino Gallo
- Interdisciplinary Center for Gene Therapy, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Tâmisa Seeko Bandeira Honda
- Interdisciplinary Center for Gene Therapy, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Patrícia Terra Alves
- Interdisciplinary Center for Gene Therapy, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sang Won Han
- Interdisciplinary Center for Gene Therapy, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil..
| |
Collapse
|
10
|
Chaintreuil P, Kerreneur E, Bourgoin M, Savy C, Favreau C, Robert G, Jacquel A, Auberger P. The generation, activation, and polarization of monocyte-derived macrophages in human malignancies. Front Immunol 2023; 14:1178337. [PMID: 37143666 PMCID: PMC10151765 DOI: 10.3389/fimmu.2023.1178337] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/04/2023] [Indexed: 05/06/2023] Open
Abstract
Macrophages are immune cells that originate from embryogenesis or from the differentiation of monocytes. They can adopt numerous phenotypes depending on their origin, tissue distribution and in response to different stimuli and tissue environment. Thus, in vivo, macrophages are endowed with a continuum of phenotypes that are rarely strictly pro-inflammatory or anti-inflammatory and exhibit a broad expression profile that sweeps over the whole polarization spectrum. Schematically, three main macrophage subpopulations coexist in human tissues: naïve macrophages also called M0, pro-inflammatory macrophages referred as M1 macrophages, and anti-inflammatory macrophages also known as M2 macrophages. Naïve macrophages display phagocytic functions, recognize pathogenic agents, and rapidly undergo polarization towards pro or anti-inflammatory macrophages to acquire their full panel of functions. Pro-inflammatory macrophages are widely involved in inflammatory response, during which they exert anti-microbial and anti-tumoral functions. By contrast, anti-inflammatory macrophages are implicated in the resolution of inflammation, the phagocytosis of cell debris and tissue reparation following injuries. Macrophages also play important deleterious or beneficial roles in the initiation and progression of different pathophysiological settings including solid and hematopoietic cancers. A better understanding of the molecular mechanisms involved in the generation, activation and polarization of macrophages is a prerequisite for the development of new therapeutic strategies to modulate macrophages functions in pathological situations.
Collapse
Affiliation(s)
- Paul Chaintreuil
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Emeline Kerreneur
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Maxence Bourgoin
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Coline Savy
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Cécile Favreau
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Guillaume Robert
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Arnaud Jacquel
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- *Correspondence: Arnaud Jacquel, ; Patrick Auberger,
| | - Patrick Auberger
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- *Correspondence: Arnaud Jacquel, ; Patrick Auberger,
| |
Collapse
|
11
|
Wang J, Liu YM, Hu J, Chen C. Trained immunity in monocyte/macrophage: Novel mechanism of phytochemicals in the treatment of atherosclerotic cardiovascular disease. Front Pharmacol 2023; 14:1109576. [PMID: 36895942 PMCID: PMC9989041 DOI: 10.3389/fphar.2023.1109576] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023] Open
Abstract
Atherosclerosis (AS) is the pathology of atherosclerotic cardiovascular diseases (ASCVD), characterized by persistent chronic inflammation in the vessel wall, in which monocytes/macrophages play a key role. It has been reported that innate immune system cells can assume a persistent proinflammatory state after short stimulation with endogenous atherogenic stimuli. The pathogenesis of AS can be influenced by this persistent hyperactivation of the innate immune system, which is termed trained immunity. Trained immunity has also been implicated as a key pathological mechanism, leading to persistent chronic inflammation in AS. Trained immunity is mediated via epigenetic and metabolic reprogramming and occurs in mature innate immune cells and their bone marrow progenitors. Natural products are promising candidates for novel pharmacological agents that can be used to prevent or treat cardiovascular diseases (CVD). A variety of natural products and agents exhibiting antiatherosclerotic abilities have been reported to potentially interfere with the pharmacological targets of trained immunity. This review describes in as much detail as possible the mechanisms involved in trained immunity and how phytochemicals of this process inhibit AS by affecting trained monocytes/macrophages.
Collapse
Affiliation(s)
- Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Yong-Mei Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| |
Collapse
|
12
|
Han J, Chitu V, Stanley ER, Wszolek ZK, Karrenbauer VD, Harris RA. Inhibition of colony stimulating factor-1 receptor (CSF-1R) as a potential therapeutic strategy for neurodegenerative diseases: opportunities and challenges. Cell Mol Life Sci 2022; 79:219. [PMID: 35366105 PMCID: PMC8976111 DOI: 10.1007/s00018-022-04225-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/06/2022] [Accepted: 02/26/2022] [Indexed: 12/12/2022]
Abstract
Microglia are specialized dynamic immune cells in the central nervous system (CNS) that plays a crucial role in brain homeostasis and in disease states. Persistent neuroinflammation is considered a hallmark of many neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson's disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS) and primary progressive multiple sclerosis (MS). Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its expression is significantly increased in neurodegenerative diseases. Cumulative findings have indicated that CSF-1R inhibitors can have beneficial effects in preclinical neurodegenerative disease models. Research using CSF-1R inhibitors has now been extended into non-human primates and humans. This review article summarizes the most recent advances using CSF-1R inhibitors in different neurodegenerative conditions including AD, PD, HD, ALS and MS. Potential challenges for translating these findings into clinical practice are presented.
Collapse
Affiliation(s)
- Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | | | - Virginija Danylaité Karrenbauer
- Department of Clinical Neuroscience, Center for Molecular Medicine L8:04, Karolinska Institutet, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
13
|
Geiß C, Salas E, Guevara-Coto J, Régnier-Vigouroux A, Mora-Rodríguez RA. Multistability in Macrophage Activation Pathways and Metabolic Implications. Cells 2022; 11:404. [PMID: 35159214 PMCID: PMC8834178 DOI: 10.3390/cells11030404] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/22/2022] Open
Abstract
Macrophages are innate immune cells with a dynamic range of reversible activation states including the classical pro-inflammatory (M1) and alternative anti-inflammatory (M2) states. Deciphering how macrophages regulate their transition from one state to the other is key for a deeper understanding of inflammatory diseases and relevant therapies. Common regulatory motifs reported for macrophage transitions, such as positive or double-negative feedback loops, exhibit a switchlike behavior, suggesting the bistability of the system. In this review, we explore the evidence for multistability (including bistability) in macrophage activation pathways at four molecular levels. First, a decision-making module in signal transduction includes mutual inhibitory interactions between M1 (STAT1, NF-KB/p50-p65) and M2 (STAT3, NF-KB/p50-p50) signaling pathways. Second, a switchlike behavior at the gene expression level includes complex network motifs of transcription factors and miRNAs. Third, these changes impact metabolic gene expression, leading to switches in energy production, NADPH and ROS production, TCA cycle functionality, biosynthesis, and nitrogen metabolism. Fourth, metabolic changes are monitored by metabolic sensors coupled to AMPK and mTOR activity to provide stability by maintaining signals promoting M1 or M2 activation. In conclusion, we identify bistability hubs as promising therapeutic targets for reverting or blocking macrophage transitions through modulation of the metabolic environment.
Collapse
Affiliation(s)
- Carsten Geiß
- Institute for Developmental Biology and Neurobiology (IDN), Johannes Gutenberg University, 55128 Mainz, Germany;
| | - Elvira Salas
- Department of Biochemistry, Faculty of Medicine, Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica;
| | - Jose Guevara-Coto
- Department of Computer Sciences and Informatics (ECCI), Faculty of Engineering, Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica;
- Research Center for Information and Communication Technologies (CITIC), Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica
| | - Anne Régnier-Vigouroux
- Institute for Developmental Biology and Neurobiology (IDN), Johannes Gutenberg University, 55128 Mainz, Germany;
| | - Rodrigo A. Mora-Rodríguez
- Institute for Developmental Biology and Neurobiology (IDN), Johannes Gutenberg University, 55128 Mainz, Germany;
- Research Center on Surgery and Cancer (CICICA), Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica
- Research Center for Tropical Diseases (CIET), Lab of Tumor Chemosensitivity (LQT), Faculty of Microbiology, Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica
| |
Collapse
|
14
|
Hastreiter AA, Paredes LC, Saraiva Camara NO. Metabolic requirement for macrophages. MACROPHAGES IN THE HUMAN BODY 2022:49-66. [DOI: 10.1016/b978-0-12-821385-8.00010-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
Hughes LD, Wang Y, Meli AP, Rothlin CV, Ghosh S. Decoding Cell Death: From a Veritable Library of Babel to Vade Mecum? Annu Rev Immunol 2021; 39:791-817. [PMID: 33902311 DOI: 10.1146/annurev-immunol-102819-072601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Programmed cell death (PCD) is a requisite feature of development and homeostasis but can also be indicative of infections, injuries, and pathologies. In concordance with these heterogeneous contexts, an array of disparate effector responses occur downstream of cell death and its clearance-spanning tissue morphogenesis, homeostatic turnover, host defense, active dampening of inflammation, and tissue repair. This raises a fundamental question of how a single contextually appropriate response ensues after an event of PCD. To explore how complex inputs may together tailor the specificity of the resulting effector response, here we consider (a) the varying contexts during which different cell death modalities are observed, (b) the nature of the information that can be passed on by cell corpses, and (c) the ways by which efferocyte populations synthesize signals from dying cells with those from the surrounding microenvironment.
Collapse
Affiliation(s)
- Lindsey D Hughes
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; , , ,
| | - Yaqiu Wang
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; , , ,
| | - Alexandre P Meli
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; , , ,
| | - Carla V Rothlin
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; , , , .,Department of Pharmacology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA;
| | - Sourav Ghosh
- Department of Pharmacology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; .,Department of Neurology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA
| |
Collapse
|
16
|
Beirão BCB, Raposo TP, Imamura LM, Ingberman M, Hupp T, Vojtěšek B, Argyle DJ. A blocking antibody against canine CSF-1R maturated by limited CDR mutagenesis. Antib Ther 2020; 3:193-204. [PMID: 33937625 PMCID: PMC7990251 DOI: 10.1093/abt/tbaa018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/02/2020] [Accepted: 07/29/2020] [Indexed: 01/15/2023] Open
Abstract
CSF-1R is a receptor mostly associated with the mononuclear phagocytic system. However, its expression within tumors has been linked with poor prognosis in both humans and dogs. Accordingly, several reports have demonstrated the beneficial effects of blocking CSF-1R in model systems of cancer. In this study, we generated a monoclonal antibody that could block CSF-1R in dogs as the first step to develop an anticancer drug for this species. Initially, an antibody was raised by the hybridoma methodology against the fragment responsible for receptor dimerization. mAb3.1, one of the resulting hybridoma clones, was able to bind macrophages in fixed tissues and was shown to inhibit cells of the mononuclear phagocytic line. Nevertheless, mAb 3.1 could not bind to some glycoforms of the receptor in its native form, while also demonstrating cross-reactivity with other proteins. To enhance binding properties of the mAb, five amino acids of the complementarity-determining region 2 of the variable heavy chain of mAb3.1 were mutated by PCR, and the variant scFv clones were screened by phage display. The selected scFv clones demonstrated improved binding to the native receptor as well as increased anti-macrophage activity. The resulting scFv antibody fragment presented here has the potential for use in cancer patients and in inflammatory diseases. Furthermore, this work provides insights into the use of such restricted mutations in antibody engineering.
Collapse
Affiliation(s)
- Breno C B Beirão
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh-Easter Bush, Midlothian, EH25 9RG, UK
| | - Teresa P Raposo
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh-Easter Bush, Midlothian, EH25 9RG, UK
| | - Louise M Imamura
- Department of Research and Development, Imunova Análises Biológicas, Curitiba, PR 80215-182, Brazil
| | - Max Ingberman
- Department of Research and Development, Imunova Análises Biológicas, Curitiba, PR 80215-182, Brazil
| | - Ted Hupp
- Cancer Research UK Edinburgh Centre MRC Institute of Genetics & Molecular Medicine, Western General Hospital, The University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Bořivoj Vojtěšek
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, 656 53, Czech Republic
| | - David J Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh-Easter Bush, Midlothian, EH25 9RG, UK
| |
Collapse
|
17
|
Human Cytomegalovirus Glycoprotein-Initiated Signaling Mediates the Aberrant Activation of Akt. J Virol 2020; 94:JVI.00167-20. [PMID: 32493823 DOI: 10.1128/jvi.00167-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/27/2020] [Indexed: 11/20/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a major cause of morbidity and mortality among immunocompromised and immunonaive individuals. HCMV-induced signaling initiated during viral entry stimulates a rapid noncanonical activation of Akt to drive the differentiation of short-lived monocytes into long-lived macrophages, which is essential for viral dissemination and persistence. We found that HCMV glycoproteins gB and gH directly bind and activate cellular epidermal growth factor receptor (EGFR) and integrin β1, respectively, to reshape canonical Akt signaling within monocytes. The remodeling of the Akt signaling network was due to the recruitment of nontraditional Akt activators to either the gB- or gH-generated receptor signaling complexes. Phosphoinositide 3-kinase (PI3K) comprised of the p110β catalytic subunit was recruited to the gB/EGFR complex despite p110δ being the primary PI3K isoform found within monocytes. Concomitantly, SH2 domain-containing inositol 5-phosphatase 1 (SHIP1) was recruited to the gH/integrin β1 complex, which is critical to aberrant Akt activation, as SHIP1 diverts PI3K signaling toward a noncanonical pathway. Although integrin β1 was required for SHIP1 recruitment, gB-activated EGFR mediated SHIP1 activation, underscoring the importance of the interplay between gB- and gH-mediated signaling to the unique activation of Akt during HCMV infection. Indeed, SHIP1 activation mediated the increased expression of Mcl-1 and HSP27, two Akt-dependent antiapoptotic proteins specifically upregulated during HCMV infection but not during growth factor treatment. Overall, our data indicate that HCMV glycoproteins gB and gH work in concert to initiate an HCMV-specific signalosome responsible for the atypical activation of Akt required for infected monocyte survival and ultimately viral persistence.IMPORTANCE Human cytomegalovirus (HCMV) infection is endemic throughout the world regardless of socioeconomic conditions and geographic locations with a seroprevalence reaching up to 100% in some developing countries. Although asymptomatic in healthy individuals, HCMV can cause severe multiorgan disease in immunocompromised or immunonaive patients. HCMV disease is a direct consequence of monocyte-mediated systematic spread of the virus following infection. Because monocytes are short-lived cells, HCMV must subvert the natural short life-span of these blood cells by inducing a distinct activation of Akt, a serine/theonine protein kinase. In this work, we demonstrate that HCMV glycoproteins gB and gH work in tandem to reroute classical host cellular receptor signaling to aberrantly activate Akt and drive survival of infected monocytes. Deciphering how HCMV modulates the cellular pathway to induce monocyte survival is important to develop a new class of anti-HCMV drugs that could target and prevent spread of the virus by eliminating infected monocytes.
Collapse
|
18
|
Lohrmann F, Forde AJ, Merck P, Henneke P. Control of myeloid cell density in barrier tissues. FEBS J 2020; 288:405-426. [PMID: 32502309 DOI: 10.1111/febs.15436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/21/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022]
Abstract
The interface between the mammalian host and its environment is formed by barrier tissues, for example, of the skin, and the respiratory and the intestinal tracts. On the one hand, barrier tissues are colonized by site-adapted microbial communities, and on the other hand, they contain specific myeloid cell networks comprising macrophages, dendritic cells, and granulocytes. These immune cells are tightly regulated in function and cell number, indicating important roles in maintaining tissue homeostasis and immune balance in the presence of commensal microorganisms. The regulation of myeloid cell density and activation involves cell-autonomous 'single-loop circuits' including autocrine mechanisms. However, an array of microenvironmental factors originating from nonimmune cells and the microbiota, as well as the microanatomical structure, impose additional layers of regulation onto resident myeloid cells. This review discusses models integrating these factors into cell-specific programs to instruct differentiation and proliferation best suited for the maintenance and renewal of immune homeostasis in the tissue-specific environment.
Collapse
Affiliation(s)
- Florens Lohrmann
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Germany.,Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany.,Spemann Graduate School for Biology and Medicine, University of Freiburg, Germany.,IMM-PACT Clinician Scientist Program, Faculty of Medicine, University of Freiburg, Germany
| | - Aaron J Forde
- Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany.,Faculty of Biology, university of Freiburg, Germany
| | - Philipp Merck
- Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany
| | - Philipp Henneke
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Germany.,Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany
| |
Collapse
|
19
|
Human Cytomegalovirus Mediates Unique Monocyte-to-Macrophage Differentiation through the PI3K/SHIP1/Akt Signaling Network. Viruses 2020; 12:v12060652. [PMID: 32560319 PMCID: PMC7354488 DOI: 10.3390/v12060652] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/29/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Blood monocytes mediate the hematogenous dissemination of human cytomegalovirus (HCMV) in the host. However, monocytes have a short 48-hour (h) lifespan and are not permissive for viral replication. We previously established that HCMV infection drives differentiation of monocytes into long-lived macrophages to mediate viral dissemination, though the mechanism was unclear. Here, we found that HCMV infection promoted monocyte polarization into distinct macrophages by inducing select M1 and M2 differentiation markers and that Akt played a central role in driving differentiation. Akt's upstream positive regulators, PI3K and SHIP1, facilitated the expression of the M1/M2 differentiation markers with p110δ being the predominant PI3K isoform inducing differentiation. Downstream of Akt, M1/M2 differentiation was mediated by caspase 3, whose activity was tightly regulated by Akt in a temporal manner. Overall, this study highlights that HCMV employs the PI3K/SHIP1/Akt pathway to regulate caspase 3 activity and drive monocyte differentiation into unique macrophages, which is critical for viral dissemination.
Collapse
|
20
|
Castegna A, Gissi R, Menga A, Montopoli M, Favia M, Viola A, Canton M. Pharmacological targets of metabolism in disease: Opportunities from macrophages. Pharmacol Ther 2020; 210:107521. [PMID: 32151665 DOI: 10.1016/j.pharmthera.2020.107521] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
From advances in the knowledge of the immune system, it is emerging that the specialized functions displayed by macrophages during the course of an immune response are supported by specific and dynamically-connected metabolic programs. The study of immunometabolism is demonstrating that metabolic adaptations play a critical role in modulating inflammation and, conversely, inflammation deeply influences the acquisition of specific metabolic settings.This strict connection has been proven to be crucial for the execution of defined immune functional programs and it is now under investigation with respect to several human disorders, such as diabetes, sepsis, cancer, and autoimmunity. The abnormal remodelling of the metabolic pathways in macrophages is now emerging as both marker of disease and potential target of therapeutic intervention. By focusing on key pathological conditions, namely obesity and diabetes, rheumatoid arthritis, atherosclerosis and cancer, we will review the metabolic targets suitable for therapeutic intervention in macrophages. In addition, we will discuss the major obstacles and challenges related to the development of therapeutic strategies for a pharmacological targeting of macrophage's metabolism.
Collapse
Affiliation(s)
- Alessandra Castegna
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; IBIOM-CNR, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| | - Rosanna Gissi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Alessio Menga
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy; Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Marcella Canton
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| |
Collapse
|
21
|
An J, Naruse TK, Hinohara K, Soejima Y, Sawabe M, Nakagawa Y, Kuwahara K, Kimura A. MRTF-A regulates proliferation and survival properties of pro-atherogenic macrophages. J Mol Cell Cardiol 2019; 133:26-35. [DOI: 10.1016/j.yjmcc.2019.05.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/01/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022]
|
22
|
Viola A, Munari F, Sánchez-Rodríguez R, Scolaro T, Castegna A. The Metabolic Signature of Macrophage Responses. Front Immunol 2019; 10:1462. [PMID: 31333642 PMCID: PMC6618143 DOI: 10.3389/fimmu.2019.01462] [Citation(s) in RCA: 1170] [Impact Index Per Article: 195.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/10/2019] [Indexed: 12/18/2022] Open
Abstract
Macrophages are a heterogeneous population of immune cells playing several and diverse functions in homeostatic and immune responses. The broad spectrum of macrophage functions depends on both heterogeneity and plasticity of these cells, which are highly specialized in sensing the microenvironment and modify their properties accordingly. Although it is clear that macrophage phenotypes are difficult to categorize and should be seen as plastic and adaptable, they can be simplified into two extremes: a pro-inflammatory (M1) and an anti-inflammatory/pro-resolving (M2) profile. Based on this definition, M1 macrophages are able to start and sustain inflammatory responses, secreting pro-inflammatory cytokines, activating endothelial cells, and inducing the recruitment of other immune cells into the inflamed tissue; on the other hand, M2 macrophages promote the resolution of inflammation, phagocytose apoptotic cells, drive collagen deposition, coordinate tissue integrity, and release anti-inflammatory mediators. Dramatic switches in cell metabolism accompany these phenotypic and functional changes of macrophages. In particular, M1 macrophages rely mainly on glycolysis and present two breaks on the TCA cycle that result in accumulation of itaconate (a microbicide compound) and succinate. Excess of succinate leads to Hypoxia Inducible Factor 1α (HIF1α) stabilization that, in turn, activates the transcription of glycolytic genes, thus sustaining the glycolytic metabolism of M1 macrophages. On the contrary, M2 cells are more dependent on oxidative phosphorylation (OXPHOS), their TCA cycle is intact and provides the substrates for the complexes of the electron transport chain (ETC). Moreover, pro- and anti-inflammatory macrophages are characterized by specific pathways that regulate the metabolism of lipids and amino acids and affect their responses. All these metabolic adaptations are functional to support macrophage activities as well as to sustain their polarization in specific contexts. The aim of this review is to discuss recent findings linking macrophage functions and metabolism.
Collapse
Affiliation(s)
- Antonella Viola
- Department of Biomedical Sciences, Istituto di Ricerca Pediatrica, University of Padova, Fondazione Città della Speranza, Padova, Italy
| | - Fabio Munari
- Department of Biomedical Sciences, Istituto di Ricerca Pediatrica, University of Padova, Fondazione Città della Speranza, Padova, Italy
| | - Ricardo Sánchez-Rodríguez
- Department of Biomedical Sciences, Istituto di Ricerca Pediatrica, University of Padova, Fondazione Città della Speranza, Padova, Italy
| | - Tommaso Scolaro
- Department of Biomedical Sciences, Istituto di Ricerca Pediatrica, University of Padova, Fondazione Città della Speranza, Padova, Italy
| | - Alessandra Castegna
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy.,IBIOM-CNR, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| |
Collapse
|
23
|
Chen HM, van der Touw W, Wang YS, Kang K, Mai S, Zhang J, Alsina-Beauchamp D, Duty JA, Mungamuri SK, Zhang B, Moran T, Flavell R, Aaronson S, Hu HM, Arase H, Ramanathan S, Flores R, Pan PY, Chen SH. Blocking immunoinhibitory receptor LILRB2 reprograms tumor-associated myeloid cells and promotes antitumor immunity. J Clin Invest 2018; 128:5647-5662. [PMID: 30352428 DOI: 10.1172/jci97570] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
Tumor-associated myeloid cells maintain immunosuppressive microenvironments within tumors. Identification of myeloid-specific receptors to modulate tumor-associated macrophage and myeloid-derived suppressor cell (MDSC) functions remains challenging. The leukocyte immunoglobulin-like receptor B (LILRB) family members are negative regulators of myeloid cell activation. We investigated how LILRB targeting could modulate tumor-associated myeloid cell function. LILRB2 antagonism inhibited receptor-mediated activation of SHP1/2 and enhanced proinflammatory responses. LILRB2 antagonism also inhibited AKT and STAT6 activation in the presence of M-CSF and IL-4. Transcriptome analysis revealed that LILRB2 antagonism altered genes involved in cell cytoskeleton remodeling, lipid/cholesterol metabolism, and endosomal sorting pathways, as well as changed differentiation gene networks associated with inflammatory myeloid cells as opposed to their alternatively activated phenotype. LILRB2 blockade effectively suppressed granulocytic MDSC and Treg infiltration and significantly promoted in vivo antitumor effects of T cell immune checkpoint inhibitors. Furthermore, LILRB2 blockade polarized tumor-infiltrating myeloid cells from non-small cell lung carcinoma tumor tissues toward an inflammatory phenotype. Our studies suggest that LILRB2 can potentially act as a myeloid immune checkpoint by reprogramming tumor-associated myeloid cells and provoking antitumor immunity.
Collapse
Affiliation(s)
- Hui-Ming Chen
- Immunotherapy Research Center, and.,Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | | | | | - Kyeongah Kang
- Immunotherapy Research Center, and.,Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | | | - Jilu Zhang
- Immunotherapy Research Center, and.,Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | | | | | | | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Richard Flavell
- Department of Immunobiology, Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Hong-Ming Hu
- Laboratory of Cancer Immunobiology, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, Oregon, USA
| | - Hisashi Arase
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Suresh Ramanathan
- Department of Thoracic Surgery, Mount Sinai Hospital, New York, New York, USA
| | - Raja Flores
- Department of Thoracic Surgery, Mount Sinai Hospital, New York, New York, USA.,Department of General Surgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ping-Ying Pan
- Immunotherapy Research Center, and.,Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Shu-Hsia Chen
- Immunotherapy Research Center, and.,Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
24
|
Temporal Manipulation of Mitochondrial Function by Virulent Francisella tularensis To Limit Inflammation and Control Cell Death. Infect Immun 2018; 86:IAI.00044-18. [PMID: 29760217 DOI: 10.1128/iai.00044-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/09/2018] [Indexed: 12/15/2022] Open
Abstract
Francisella tularensis subsp. tularensis is a highly pathogenic intracellular bacterium that suppresses host inflammation by impairing the metabolic shift from oxidative phosphorylation to glycolysis. Decreased mitochondrial metabolism is central to initiating a metabolic shift to glycolysis and regulating inflammation, but F. tularensis subsp. tularensis manipulation of host mitochondrial function has not been explored. We demonstrate, using extracellular flux analysis, that F. tularensis subsp. tularensis infection initially improves host macrophage mitochondrial bioenergetics in a capsule-dependent manner. Enhancement of mitochondrial function by F. tularensis subsp. tularensis allowed for modest replication and inhibition of apoptosis early after infection. However, using live cell imaging, we found that F. tularensis subsp. tularensis facilitated the loss of mitochondrial function at later time points during infection in a capsule-independent fashion. This loss of function was paired with oncosis and rapid bacterial replication. Inhibition of oncosis reduced intracellular bacterial numbers, underscoring the requirement for this process during F. tularensis subsp. tularensis infection. These findings establish that temporal mitochondrial manipulation by F. tularensis subsp. tularensis is critical for maintenance of a noninflammatory environment and subsequently aids in optimal replication and dissemination of this pathogenic organism.
Collapse
|
25
|
Sasaki T, Tanaka Y, Kulkeaw K, Yumine-Takai A, Tan KS, Nishinakamura R, Ishida J, Fukamizu A, Sugiyama D. Embryonic Intra-Aortic Clusters Undergo Myeloid Differentiation Mediated by Mesonephros-Derived CSF1 in Mouse. Stem Cell Rev Rep 2017; 12:530-542. [PMID: 27324145 DOI: 10.1007/s12015-016-9668-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The aorta-gonad-mesonephros (AGM) region contains intra-aortic clusters (IACs) thought to have acquired hematopoietic stem cell (HSC) potential in vertebrate embryos. To assess extrinsic regulation of IACs in the AGM region, we employed mouse embryos harboring a Sall1-GFP reporter gene, which allows identification of mesonephros cells based on GFP expression. Analysis of AGM region tissue sections confirmed mesonephros GFP expression. Mesonephric cells sorted at E10.5 expressed mRNA encoding Csf1, a hematopoietic cytokine, and corresponding protein, based on real-time PCR and immunocytochemistry, respectively. Further analysis indicated that some IACs express the CSF1 receptor, CSF1R. Expression of Cebpa and Irf8 mRNAs was higher in CSF1R-positive IACs, whereas that of Cebpε and Gfi1 mRNAs was lower relative to CSF1R-negative IACs, suggesting that CSF1/CSF1R signaling functions in IAC myeloid differentiation by modulating expression of these transcription factors. Colony formation assays using CSF1R-positive IACs revealed increased numbers of myeloid colonies in the presence of CSF1. Analysis using an intra-cellular signaling array indicated the greatest fold increase of Cleaved Caspase-3 in AGM cells in the presence of CSF1. Immunohistochemistry revealed that Cleaved Caspase-3 is primarily expressed in IACs in the AGM region, and incubation of IACs with CSF1 up-regulated Cleaved Caspase-3. Overall, our findings suggest that CSF1 secreted from mesonephros accelerates IAC myeloid differentiation in the AGM region, possibly via Caspase-3 cleavage.
Collapse
Affiliation(s)
- Tatsuya Sasaki
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
| | - Yuka Tanaka
- Department of Research and Development of Next Generation Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Kasem Kulkeaw
- Department of Research and Development of Next Generation Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ayako Yumine-Takai
- Department of Research and Development of Next Generation Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keai Sinn Tan
- Department of Research and Development of Next Generation Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Junji Ishida
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
- Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
| | - Akiyoshi Fukamizu
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
- Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
| | - Daisuke Sugiyama
- Department of Research and Development of Next Generation Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
- Center for Clinical and Translational Research, Kyushu University, Fukuoka, 812-84582, Japan.
- Department of Clinical Study, Center for Advanced Medical Innovation, Kyushu University, Station for Collaborative Research 1 4F, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
26
|
Llewellyn RA, Thomas KS, Gutknecht MF, Bouton AH. The nonreceptor protein tyrosine kinase Pyk2 promotes the turnover of monocytes at steady state. J Leukoc Biol 2017; 102:1069-1080. [PMID: 28754799 DOI: 10.1189/jlb.1a0217-063r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/20/2017] [Accepted: 07/10/2017] [Indexed: 12/30/2022] Open
Abstract
Monocytes are short-lived myeloid cells that perform functions essential for tissue homeostasis and disease resolution. However, the cellular mechanisms controlling the maintenance and turnover of monocyte populations are largely undefined. Proline-rich tyrosine kinase 2 (Pyk2) is a nonreceptor tyrosine kinase that regulates numerous immune cell functions, but its role in monocytes is currently unknown. In this study, we sought to characterize the expression and function of Pyk2 in lineage-committed monocyte populations. Here, we report that Pyk2 protein expression is increased in the Ly6C- monocyte population. Using a Pyk2 knockout mouse model (Pyk2-/-), we show that Pyk2 regulates the relative proportion of monocyte subsets normally represented in the bone marrow (BM) at steady state. In support of this conclusion, a similar phenotype was observed in the peripheral blood and spleen. Data from reciprocal BM chimera experiments indicate that the alterations in monocyte populations exhibited by Pyk2-/- mice are due to factors intrinsic to the monocytes. Lineage-tracing of monocyte populations suggests that Pyk2 promotes apoptosis in BM monocytes, thereby acting as an important homeostatic regulator of turnover in these short-lived, innate immune cells.
Collapse
Affiliation(s)
- Ryan A Llewellyn
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Keena S Thomas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Michael F Gutknecht
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Amy H Bouton
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
27
|
Bellora F, Dondero A, Corrias MV, Casu B, Regis S, Caliendo F, Moretta A, Cazzola M, Elena C, Vinti L, Locatelli F, Bottino C, Castriconi R. Imatinib and Nilotinib Off-Target Effects on Human NK Cells, Monocytes, and M2 Macrophages. THE JOURNAL OF IMMUNOLOGY 2017; 199:1516-1525. [PMID: 28701512 DOI: 10.4049/jimmunol.1601695] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 06/11/2017] [Indexed: 12/22/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are used in the clinical management of hematological neoplasms. Moreover, in solid tumors such as stage 4 neuroblastomas (NB), imatinib showed benefits that might depend on both on-target and immunological off-target effects. We investigated the effects of imatinib and nilotinib on human NK cells, monocytes, and macrophages. High numbers of monocytes died upon exposure to TKI concentrations similar to those achieved in patients. Conversely, NK cells were highly resistant to the TKI cytotoxic effect, were properly activated by immunostimulatory cytokines, and degranulated in the presence of NB cells. In NB, neither drug reduced the expression of ligands for activating NK receptors or upregulated that of HLA class I, B7-H3, PD-L1, and PD-L2, molecules that might limit NK cell function. Interestingly, TKIs modulated the chemokine receptor repertoire of immune cells. Acting at the transcriptional level, they increased the surface expression of CXCR4, an effect observed also in NK cells and monocytes of patients receiving imatinib for chronic myeloid leukemia. Moreover, TKIs reduced the expression of CXCR3 (in NK cells) and CCR1 (in monocytes). Monocytes also decreased the expression of M-CSFR, and low numbers of cells underwent differentiation toward macrophages. M0 and M2 macrophages were highly resistant to TKIs and maintained their phenotypic and functional characteristics. Importantly, also in the presence of TKIs, the M2 immunosuppressive polarization was reverted by TLR engagement, and M1-oriented macrophages fully activated autologous NK cells. Our results contribute to better interpreting the off-target efficacy of TKIs in tumors and to envisaging strategies aimed at facilitating antitumor immune responses.
Collapse
Affiliation(s)
- Francesca Bellora
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, 16132 Genoa, Italy
| | - Alessandra Dondero
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, 16132 Genoa, Italy
| | | | - Beatrice Casu
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, 16132 Genoa, Italy
| | | | - Fabio Caliendo
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, 16132 Genoa, Italy
| | - Alessandro Moretta
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, 16132 Genoa, Italy; .,Centro di Eccellenza per la Ricerca Biomedica, Università degli Studi di Genova, 16132 Genoa, Italy
| | - Mario Cazzola
- Dipartimento di Medicina Molecolare, Università di Pavia, 27100 Pavia, Italy.,Dipartimento di Onco-Ematologia, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, 27100 Pavia, Italy; and
| | - Chiara Elena
- Dipartimento di Medicina Molecolare, Università di Pavia, 27100 Pavia, Italy
| | - Luciana Vinti
- Dipartimento di Onco-Ematologia Pediatrica, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy
| | - Franco Locatelli
- Dipartimento di Medicina Molecolare, Università di Pavia, 27100 Pavia, Italy.,Dipartimento di Onco-Ematologia Pediatrica, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy
| | - Cristina Bottino
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, 16132 Genoa, Italy.,Istituto Giannina Gaslini, 16148 Genoa, Italy
| | - Roberta Castriconi
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, 16132 Genoa, Italy.,Centro di Eccellenza per la Ricerca Biomedica, Università degli Studi di Genova, 16132 Genoa, Italy
| |
Collapse
|
28
|
Wilkins HM, Koppel SJ, Weidling IW, Roy N, Ryan LN, Stanford JA, Swerdlow RH. Extracellular Mitochondria and Mitochondrial Components Act as Damage-Associated Molecular Pattern Molecules in the Mouse Brain. J Neuroimmune Pharmacol 2016; 11:622-628. [PMID: 27562848 DOI: 10.1007/s11481-016-9704-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/17/2016] [Indexed: 11/26/2022]
Abstract
Mitochondria and mitochondrial debris are found in the brain's extracellular space, and extracellular mitochondrial components can act as damage associated molecular pattern (DAMP) molecules. To characterize the effects of potential mitochondrial DAMP molecules on neuroinflammation, we injected either isolated mitochondria or mitochondrial DNA (mtDNA) into hippocampi of C57BL/6 mice and seven days later measured markers of inflammation. Brains injected with whole mitochondria showed increased Tnfα and decreased Trem2 mRNA, increased GFAP protein, and increased NFκB phosphorylation. Some of these effects were also observed in brains injected with mtDNA (decreased Trem2 mRNA, increased GFAP protein, and increased NFκB phosphorylation), and mtDNA injection also caused several unique changes including increased CSF1R protein and AKT phosphorylation. To further establish the potential relevance of this response to Alzheimer's disease (AD), a brain disorder characterized by neurodegeneration, mitochondrial dysfunction, and neuroinflammation we also measured App mRNA, APP protein, and Aβ1-42 levels. We found mitochondria (but not mtDNA) injections increased these parameters. Our data show that in the mouse brain extracellular mitochondria and its components can induce neuroinflammation, extracellular mtDNA or mtDNA-associated proteins can contribute to this effect, and mitochondria derived-DAMP molecules can influence AD-associated biomarkers.
Collapse
Affiliation(s)
- Heather M Wilkins
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA
| | - Scott J Koppel
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA
| | - Ian W Weidling
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nairita Roy
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lauren N Ryan
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA
| | - John A Stanford
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA.
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.
- University of Kansas School of Medicine, MS 2012, Landon Center on Aging, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| |
Collapse
|
29
|
Colony-Stimulating Factor 1 Receptor Antagonists Sensitize Human Immunodeficiency Virus Type 1-Infected Macrophages to TRAIL-Mediated Killing. J Virol 2016; 90:6255-6262. [PMID: 27122585 DOI: 10.1128/jvi.00231-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/21/2016] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED Strategies aimed at eliminating persistent viral reservoirs from HIV-1-infected individuals have focused on CD4(+) T-cell reservoirs. However, very little attention has been given to approaches that could promote elimination of tissue macrophage reservoirs. HIV-1 infection of macrophages induces phosphorylation of colony-stimulating factor 1 receptor (CSF-1R), which confers resistance to apoptotic pathways driven by tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), thereby promoting viral persistence. In this study, we assessed whether CSF-1R antagonists (PLX647, PLX3397, and PLX5622) restored apoptotic sensitivity of HIV-1-infected macrophages in vitro PLX647, PLX3397, and PLX5622 at clinically relevant concentrations blocked the activation of CSF-1R and reduced the viability of infected macrophages, as well as the extent of viral replication. Our data show that strategies targeting monocyte colony-stimulating factor (MCSF) signaling could be used to promote elimination of HIV-1-infected myeloid cells and to contribute to the elimination of persistent viral reservoirs. IMPORTANCE As the HIV/AIDS research field explores approaches to eliminate HIV-1 in individuals on suppressive antiviral therapy, those approaches will need to eliminate both CD4(+) T-cell and myeloid cell reservoirs. Most of the attention has focused on CD4(+) T-cell reservoirs, and scant attention has been paid to myeloid cell reservoirs. The distinct nature of the infection in myeloid cells versus CD4(+) T cells will likely dictate different approaches in order to achieve their elimination. For CD4(+) T cells, most strategies focus on promoting virus reactivation to promote immune-mediated clearance and/or elimination by viral cytopathicity. Macrophages resist viral cytopathic effects and CD8(+) T-cell killing. Therefore, we have explored clearance strategies that render macrophages sensitive to viral cytopathicity. This research helps inform the design of strategies to promote clearance of the macrophage reservoir in infected individuals on suppressive antiviral therapy.
Collapse
|
30
|
Human Cytomegalovirus Induces an Atypical Activation of Akt To Stimulate the Survival of Short-Lived Monocytes. J Virol 2016; 90:6443-6452. [PMID: 27147739 DOI: 10.1128/jvi.00214-16] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/26/2016] [Indexed: 02/02/2023] Open
Abstract
UNLABELLED Human cytomegalovirus (HCMV) is a pervasive herpesvirus responsible for significant morbidity and mortality among immunodeficient/naive hosts. Following a primary HCMV infection, circulating blood monocytes mediate the systemic spread of the virus. Extending the short 48-h life span of monocytes is critical to the viral dissemination process, as these blood-borne cells are nonpermissive for virus replication until they are fully differentiated into macrophages. Here, we show that HCMV glycoprotein gB binding to cellular epidermal growth factor receptor (EGFR) during HCMV entry initiated a rapid (within 15 min) activation of the apoptosis suppressor Akt, which was maintained through 72 h. The virus-induced activation of Akt was more robust than that with the normal myeloid growth factor macrophage colony-stimulating factor (M-CSF) and was essential for infected monocytes to bypass the 48-h viability checkpoint. Activation of phosphoinositide 3-kinase (PI3K) following EGFR engagement by HCMV mediated the phosphorylation of Akt. Moreover, HCMV entry drove a switch away from the PI3K p110δ isoform, which was required for the viability of uninfected monocytes, to the p110β isoform in order to facilitate the Akt-dependent prosurvival state within infected cells. Simultaneously, in contrast to M-CSF, HCMV promoted a rapid increase in SH2 domain-containing inositol 5-phosphatase 1 (SHIP1) expression, leading to signaling through a noncanonical Akt activation pathway. To ensure maximum Akt activity, HCMV also induced an early phosphorylation-dependent inactivation of the negative regulator phosphatase and tensin homolog. Overall, our data indicate that HCMV hijacks the upstream Akt signaling network to induce a nontraditional activation of Akt and subsequently a prosurvival decision at the 48-h cell fate checkpoint, a vital step for HCMV's dissemination and persistence strategy. IMPORTANCE HCMV is found throughout the world with a prevalence of 55 to 100% within the human population. HCMV infection is generally asymptomatic in immunocompetent or naive individuals but is a significant cause of morbidity and mortality among the immunocompromised. Widespread organ inflammation is associated with symptomatic infections, which is a direct consequence of the viral dissemination strategy. Inflammatory peripheral blood monocytes facilitate the spread of HCMV. However, HCMV must subvert the naturally short life span of monocytes. In this work, we demonstrate that HCMV induces the activation of Akt, an antiapoptotic protein, in a manner distinct from that of normal myeloid growth factors. Moreover, we decipher how HCMV dysregulates the upstream Akt signaling network during viral entry to promote an Akt-dependent prosurvival state following infection. Delineation of the virus-specific mechanisms that regulate cellular prosurvival pathways in order to drive the survival of HCMV-infected monocytes is important to identifying new anti-HCMV therapeutic targets.
Collapse
|
31
|
Kim HJ, Ohk B, Kang WY, Seong SJ, Suk K, Lim MS, Kim SY, Yoon YR. Deficiency of Lipocalin-2 Promotes Proliferation and Differentiation of Osteoclast Precursors via Regulation of c-Fms Expression and Nuclear Factor-kappa B Activation. J Bone Metab 2016; 23:8-15. [PMID: 26981515 PMCID: PMC4791440 DOI: 10.11005/jbm.2016.23.1.8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/02/2016] [Accepted: 02/02/2016] [Indexed: 01/27/2023] Open
Abstract
Background Lipocalin-2 (LCN2), a small glycoprotein, has a pivotal role in diverse biological processes such as cellular proliferation and differentiation. We previously reported that LCN2 is implicated in osteoclast formation induced by receptor activator of nuclear factor-kappa B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). In the present study, we used a knockout mouse model to further investigate the role of LCN2 in osteoclast development. Methods Osteoclastogenesis was assessed using primary bone marrow-derived macrophages. RANKL and M-CSF signaling was determined by immunoblotting, cell proliferation by bromodeoxyuridine (BrdU) enzyme-linked immunosorbent assay (ELISA), and apoptosis by cell death detection ELISA. Bone morphometric parameters were determined using a micro-computed tomography system. Results Our results showed that LCN2 deficiency increases tartrate-resistant acid phosphatase (TRAP)-positive multinucleated osteoclast formation in vitro, a finding that reflects enhanced proliferation and differentiation of osteoclast lineage cells. LCN2 deficiency promotes M-CSF-induced proliferation of bone marrow macrophages (BMMs), osteoclast precursors, without altering their survival. The accelerated proliferation of LCN2-deficient precursors is associated with enhanced expression and activation of the M-CSF receptor, c-Fms. Furthermore, LCN2 deficiency stimulates the induction of c-Fos and nuclear factor of activated T cells c1 (NFATc1), key transcription factors for osteoclastogenesis, and promotes RANKL-induced inhibitor of kappa B (IκBα) phosphorylation. Interestingly, LCN2 deficiency does not affect basal osteoclast formation in vivo, suggesting that LCN2 might play a role in the enhanced osteoclast development that occurs under some pathological conditions. Conclusions Our study establishes LCN2 as a negative modulator of osteoclast formation, results that are in accordance with our previous findings.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea.; Skeletal Diseases Genome Research Center, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Boram Ohk
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea
| | - Woo Youl Kang
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea
| | - Sook Jin Seong
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Mi-Sun Lim
- College of Pharmacy, Yeungnam University, Gyeongsan, Korea
| | - Shin-Yoon Kim
- Skeletal Diseases Genome Research Center, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Young-Ran Yoon
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu, Korea
| |
Collapse
|
32
|
Song X, Ding Y, Liu G, Yang X, Zhao R, Zhang Y, Zhao X, Anderson GJ, Nie G. Cancer Cell-derived Exosomes Induce Mitogen-activated Protein Kinase-dependent Monocyte Survival by Transport of Functional Receptor Tyrosine Kinases. J Biol Chem 2016; 291:8453-64. [PMID: 26895960 DOI: 10.1074/jbc.m116.716316] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Indexed: 12/31/2022] Open
Abstract
Tumor-associated macrophages (TAM) play pivotal roles in cancer initiation and progression. Monocytes, the precursors of TAMs, normally undergo spontaneous apoptosis within 2 days, but can subsist in the inflammatory tumor microenvironment for continuous survival and generation of sufficient TAMs. The mechanisms underlying tumor-driving monocyte survival remain obscure. Here we report that cancer cell-derived exosomes were crucial mediators for monocyte survival in the inflammatory niche. Analysis of the survival-promoting molecules in monocytes revealed that cancer cell-derived exosomes activated Ras and extracellular signal-regulated kinases in the mitogen-activated protein kinase (MAPK) pathway, resulting in the prevention of caspase cleavage. Phosphorylated receptor tyrosine kinases (RTKs), such as phosphorylated epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER-2), were abundantly expressed in cancer cell-derived exosomes. Knock-out of EGFR or/and HER-2, or alternatively, inhibitors against their phosphorylation significantly disturbed the exosome-mediated activation of the MAPK pathway, inhibition of caspase cleavage, and increase in survival rate in monocytes. Moreover, the deprived survival-stimulating activity of exosomes due to null expression of EGFR and HER-2 could be restored by activation of another RTK, insulin receptor. Overall, our study uncovered a mechanism of tumor-associated monocyte survival and demonstrated that cancer cell-derived exosomes can stimulate the MAPK pathway in monocytes through transport of functional RTKs, leading to inactivation of apoptosis-related caspases. This work provides insights into the long sought question on monocyte survival prior to formation of plentiful TAMs in the tumor microenvironment.
Collapse
Affiliation(s)
- Xiao Song
- From the CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11th Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Yanping Ding
- From the CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11th Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Gang Liu
- From the CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11th Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Xiao Yang
- From the CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11th Beiyitiao, Zhongguancun, Beijing 100190, China, the College of Pharmaceutical Science, Jilin University, Changchun 130021, China
| | - Ruifang Zhao
- From the CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11th Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Yinlong Zhang
- From the CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11th Beiyitiao, Zhongguancun, Beijing 100190, China, the College of Pharmaceutical Science, Jilin University, Changchun 130021, China
| | - Xiao Zhao
- From the CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11th Beiyitiao, Zhongguancun, Beijing 100190, China, the Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China, and
| | - Gregory J Anderson
- the Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Locked Bag 2000, Royal Brisbane Hospital, Brisbane, Queensland 4029, Australia
| | - Guangjun Nie
- From the CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, 11th Beiyitiao, Zhongguancun, Beijing 100190, China,
| |
Collapse
|
33
|
Kim MJ, Kim H, Lee SH, Gu DR, Lee SY, Lee K, Jeong D. ADP-Ribosylation Factor 1 Regulates Proliferation, Migration, and Fusion in Early Stage of Osteoclast Differentiation. Int J Mol Sci 2015; 16:29305-14. [PMID: 26690137 PMCID: PMC4691111 DOI: 10.3390/ijms161226168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 01/12/2023] Open
Abstract
Small G-protein adenosine diphosphate (ADP)-ribosylation factors (ARFs) regulate a variety of cellular functions, including actin cytoskeleton remodeling, plasma membrane reorganization, and vesicular transport. Here, we propose the functional roles of ARF1 in multiple stages of osteoclast differentiation. ARF1 was upregulated during receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation and transiently activated in an initial stage of their differentiation. Differentiation of ARF1-deficient osteoclast precursors into mature osteoclasts temporarily increased in pre-maturation stage of osteoclasts followed by reduced formation of mature osteoclasts, indicating that ARF1 regulates the osteoclastogenic process. ARF1 deficiency resulted in reduced osteoclast precursor proliferation and migration as well as increasing cell-cell fusion. In addition, ARF1 silencing downregulated c-Jun N-terminal kinase (JNK), Akt, osteopontin, and macrophage colony-stimulating factor (M-CSF)-receptor c-Fms as well as upregulating several fusion-related genes including CD44, CD47, E-cadherin, and meltrin-α. Collectively, we showed that ARF1 stimulated proliferation and migration of osteoclast precursors while suppressing their fusion, suggesting that ARF1 may be a plausible inter-player that mediates the transition to osteoclast fusion at multiple steps during osteoclast differentiation.
Collapse
Affiliation(s)
- Min Jae Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 705-717, Korea.
| | - Hyunsoo Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 705-717, Korea.
| | - Seoung Hoon Lee
- Department of Oral Microbiology and Immunology, College of Dentistry, Wonkwang University, Iksan 570-749, Korea.
- Center for Metabolic Function Regulation (CMFR), Wonkwang University School of Medicine, Iksan 570-749, Korea.
| | - Dong Ryun Gu
- Center for Metabolic Function Regulation (CMFR), Wonkwang University School of Medicine, Iksan 570-749, Korea.
| | - Soo Young Lee
- Department of Life Science and Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750, Korea.
| | - Kyunghee Lee
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 705-717, Korea.
| | - Daewon Jeong
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 705-717, Korea.
| |
Collapse
|
34
|
Richardson ET, Shukla S, Nagy N, Boom WH, Beck RC, Zhou L, Landreth GE, Harding CV. ERK Signaling Is Essential for Macrophage Development. PLoS One 2015; 10:e0140064. [PMID: 26445168 PMCID: PMC4596867 DOI: 10.1371/journal.pone.0140064] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/20/2015] [Indexed: 11/25/2022] Open
Abstract
Macrophages depend on colony stimulating factor 1 (also known as M-CSF) for their growth and differentiation, but the requirements for intracellular signals that lead to macrophage differentiation and function remain unclear. M-CSF is known to activate ERK1 and ERK2, but the importance of this signaling pathway in macrophage development is unknown. In these studies, we characterized a novel model of Erk1-/-Erk2flox/floxLyz2Cre/Cre mice in which the ERK2 isoform is deleted from macrophages in the background of global ERK1 deficiency. Cultures of M-CSF-stimulated bone marrow precursors from these mice yielded reduced numbers of macrophages. Whereas macrophages developing from M-CSF-stimulated bone marrow of Erk2flox/floxLyz2Cre/Cre mice showed essentially complete loss of ERK2 expression, the reduced number of macrophages that develop from Erk1-/-Erk2flox/floxLyz2Cre/Cre bone marrow show retention of ERK2 expression, indicating selective outgrowth of a small proportion of precursors in which Cre-mediated deletion failed to occur. The bone marrow of Erk1-/-Erk2flox/floxLyz2Cre/Cre mice was enriched for CD11b+ myeloid cells, CD11bhi Gr-1hi neutrophils, Lin- c-Kit+ Sca–1+ hematopoietic stem cells, and Lin- c-Kit+ CD34+ CD16/32+ granulocyte-macrophage progenitors. Culture of bone marrow Lin- cells under myeloid-stimulating conditions yielded reduced numbers of monocytes. Collectively, these data indicate that the defect in production of macrophages is not due to a reduced number of progenitors, but rather due to reduced ability of progenitors to proliferate and produce macrophages in response to M-CSF-triggered ERK signaling. Macrophages from Erk1-/-Erk2flox/floxLyz2Cre/Cre bone marrow showed reduced induction of M-CSF-regulated genes that depend on the ERK pathway for their expression. These data demonstrate that ERK1/ERK2 play a critical role in driving M-CSF-dependent proliferation of bone marrow progenitors for production of macrophages.
Collapse
Affiliation(s)
- Edward T. Richardson
- Department of Pathology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
- Medical Scientist Training Program, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Supriya Shukla
- Department of Pathology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Nancy Nagy
- Department of Pathology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - W. Henry Boom
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
- Center for AIDS Research, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Rose C. Beck
- Department of Pathology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Lan Zhou
- Department of Pathology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
| | - Gary E. Landreth
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Clifford V. Harding
- Department of Pathology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
- Center for AIDS Research, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
35
|
pGlcNAc Nanofiber Treatment of Cutaneous Wounds Stimulate Increased Tensile Strength and Reduced Scarring via Activation of Akt1. PLoS One 2015; 10:e0127876. [PMID: 25955155 PMCID: PMC4425470 DOI: 10.1371/journal.pone.0127876] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/21/2015] [Indexed: 11/30/2022] Open
Abstract
Treatment of cutaneous wounds with poly-N-acetyl-glucosamine containing nanofibers (pGlcNAc), a novel polysaccharide material derived from a marine diatom, results in increased wound closure, antibacterial activities and innate immune responses. We have shown that Akt1 plays a central role in the regulation of these activities. Here, we show that pGlcNAc treatment of cutaneous wounds results in a smaller scar that has increased tensile strength and elasticity. pGlcNAc treated wounds exhibit decreased collagen content, increased collagen organization and decreased myofibroblast content. A fibrin gel assay was used to assess the regulation of fibroblast alignment in vitro. In this assay, fibrin lattice is formed with two pins that provide focal points upon which the gel can exert force as the cells align from pole to pole. pGlcNAc stimulation of embedded fibroblasts results in cellular alignment as compared to untreated controls, by a process that is Akt1 dependent. We show that Akt1 is required in vivo for the pGlcNAc-induced increased tensile strength and elasticity. Taken together, our findings suggest that pGlcNAc nanofibers stimulate an Akt1 dependent pathway that results in the proper alignment of fibroblasts, decreased scarring, and increased tensile strength during cutaneous wound healing.
Collapse
|
36
|
Malavez Y, Voss OH, Gonzalez-Mejia ME, Parihar A, Doseff AI. Distinct contribution of protein kinase Cδ and protein kinase Cε in the lifespan and immune response of human blood monocyte subpopulations. Immunology 2015; 144:611-20. [PMID: 25322815 DOI: 10.1111/imm.12412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 02/06/2023] Open
Abstract
Monocytes, key components of the immune system, are a heterogeneous population comprised of classical monocytes (CD16(-) ) and non-classical monocytes (CD16(+) ). Monocytes are short lived and undergo spontaneous apoptosis, unless stimulated. Dysregulation of monocyte numbers contribute to the pathophysiology of inflammatory diseases, yet the contribution of each subset remains poorly characterized. Protein kinase C (PKC) family members are central to monocyte biology; however, their role in regulating lifespan and immune function of CD16(-) and CD16(+) monocytes has not been studied. Here, we evaluated the contribution of PKCδ and PKCε in the lifespan and immune response of both monocyte subsets. We showed that CD16(+) monocytes are more susceptible to spontaneous apoptosis because of the increased caspase-3, -8 and -9 activities accompanied by higher kinase activity of PKCδ. Silencing of PKCδ reduced apoptosis in both CD16(+) and CD16(-) monocytes. CD16(+) monocytes express significantly higher levels of PKCε and produce more tumour necrosis factor-α in CD16(+) compared with CD16(-) monocytes. Silencing of PKCε affected the survival and tumour necrosis factor-α production. These findings demonstrate a complex network with similar topography, yet unique regulatory characteristics controlling lifespan and immune response in each monocyte subset, helping define subset-specific coordination programmes controlling monocyte function.
Collapse
Affiliation(s)
- Yadira Malavez
- Department of Molecular Genetics, Department of Internal Medicine, Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | | | | | | | | |
Collapse
|
37
|
Cheng SC, Quintin J, Cramer RA, Shepardson KM, Saeed S, Kumar V, Giamarellos-Bourboulis EJ, Martens JHA, Rao NA, Aghajanirefah A, Manjeri GR, Li Y, Ifrim DC, Arts RJW, van der Veer BMJW, van der Meer BMJW, Deen PMT, Logie C, O'Neill LA, Willems P, van de Veerdonk FL, van der Meer JWM, Ng A, Joosten LAB, Wijmenga C, Stunnenberg HG, Xavier RJ, Netea MG. mTOR- and HIF-1α-mediated aerobic glycolysis as metabolic basis for trained immunity. Science 2014; 345:1250684. [PMID: 25258083 DOI: 10.1126/science.1250684] [Citation(s) in RCA: 1463] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epigenetic reprogramming of myeloid cells, also known as trained immunity, confers nonspecific protection from secondary infections. Using histone modification profiles of human monocytes trained with the Candida albicans cell wall constituent β-glucan, together with a genome-wide transcriptome, we identified the induced expression of genes involved in glucose metabolism. Trained monocytes display high glucose consumption, high lactate production, and a high ratio of nicotinamide adenine dinucleotide (NAD(+)) to its reduced form (NADH), reflecting a shift in metabolism with an increase in glycolysis dependent on the activation of mammalian target of rapamycin (mTOR) through a dectin-1-Akt-HIF-1α (hypoxia-inducible factor-1α) pathway. Inhibition of Akt, mTOR, or HIF-1α blocked monocyte induction of trained immunity, whereas the adenosine monophosphate-activated protein kinase activator metformin inhibited the innate immune response to fungal infection. Mice with a myeloid cell-specific defect in HIF-1α were unable to mount trained immunity against bacterial sepsis. Our results indicate that induction of aerobic glycolysis through an Akt-mTOR-HIF-1α pathway represents the metabolic basis of trained immunity.
Collapse
Affiliation(s)
- Shih-Chin Cheng
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Jessica Quintin
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Robert A Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Kelly M Shepardson
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Sadia Saeed
- Department of Molecular Biology, Faculties of Science and Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, Netherlands
| | - Vinod Kumar
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | | | - Joost H A Martens
- Department of Molecular Biology, Faculties of Science and Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, Netherlands
| | - Nagesha Appukudige Rao
- Department of Molecular Biology, Faculties of Science and Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, Netherlands
| | - Ali Aghajanirefah
- Department of Molecular Biology, Faculties of Science and Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, Netherlands
| | - Ganesh R Manjeri
- Department of Biochemistry, Faculties of Science and Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, Netherlands
| | - Yang Li
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Daniela C Ifrim
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Rob J W Arts
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Brian M J W van der Veer
- Department of Molecular Biology, Faculties of Science and Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, Netherlands
| | - Brian M J W van der Meer
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Peter M T Deen
- Department of Physiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Colin Logie
- Department of Molecular Biology, Faculties of Science and Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, Netherlands
| | - Luke A O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Peter Willems
- Department of Biochemistry, Faculties of Science and Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, Netherlands
| | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Jos W M van der Meer
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Aylwin Ng
- Center for Computational and Integrative Biology and Gastrointestinal Unit, Massachusetts General Hospital, Harvard School of Medicine, Boston, MA 02114, USA. Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Hendrik G Stunnenberg
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Ramnik J Xavier
- Center for Computational and Integrative Biology and Gastrointestinal Unit, Massachusetts General Hospital, Harvard School of Medicine, Boston, MA 02114, USA. Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands.
| |
Collapse
|
38
|
Busca A, Saxena M, Iqbal S, Angel J, Kumar A. PI3K/Akt regulates survival during differentiation of human macrophages by maintaining NF-κB-dependent expression of antiapoptotic Bcl-xL. J Leukoc Biol 2014; 96:1011-22. [DOI: 10.1189/jlb.1a0414-212r] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
39
|
Chen T, Wang X, Guo L, Wu M, Duan Z, Lv J, Tai W, Renganathan H, Didier R, Li J, Sun D, Chen X, He X, Fan J, Young W, Ren Y. Embryonic Stem Cells Promoting Macrophage Survival and Function are Crucial for Teratoma Development. Front Immunol 2014; 5:275. [PMID: 25071759 PMCID: PMC4082241 DOI: 10.3389/fimmu.2014.00275] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/27/2014] [Indexed: 11/29/2022] Open
Abstract
Stem cell therapies have had tremendous potential application for many diseases in recent years. However, the tumorigenic properties of stem cells restrict their potential clinical application; therefore, strategies for reducing the tumorigenic potential of stem cells must be established prior to transplantation. We have demonstrated that syngeneic transplantation of embryonic stem cells (ESCs) provokes an inflammatory response that involves the rapid recruitment of bone marrow-derived macrophages (BMDMs). ESCs are able to prevent mature macrophages from macrophage colony-stimulating factor (M-CSF) withdrawal-induced apoptosis, and thus prolong macrophage lifespan significantly by blocking various apoptotic pathways in an M-CSF-independent manner. ESCs express and secrete IL-34, which may be responsible for ESC-promoted macrophage survival. This anti-apoptotic effect of ESCs involves activation of extracellular signal-regulated kinase (ERK)1/2 and PI3K/Akt pathways and thus, inhibition of ERK1/2 and PI3K/AKT activation decreases ESC-induced macrophage survival. Functionally, ESC-treated macrophages also showed a higher level of phagocytic activity. ESCs further serve to polarize BMDMs into M2-like macrophages that exhibit most tumor-associated macrophage phenotypic and functional features. ESC-educated macrophages produce high levels of arginase-1, Tie-2, and TNF-α, which participate in angiogenesis and contribute to teratoma progression. Our study suggests that induction of M2-like macrophage activation is an important mechanism for teratoma development. Strategies targeting macrophages to inhibit teratoma development would increase the safety of ESC-based therapies, inasmuch as the depletion of macrophages completely inhibits ESC-induced angiogenesis and teratoma development.
Collapse
Affiliation(s)
- Tianxiang Chen
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey , New Jersey, NJ , USA ; Department of Thoracic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , China
| | - Xi Wang
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey , New Jersey, NJ , USA ; Institute of Neurosciences, The Fourth Military Medical University , Xian , China
| | - Lei Guo
- Department of Biomedical Sciences, Florida State University College of Medicine , Tallahassee, FL , USA ; Department of Orthopedic Surgery, The Second Hospital of Xian Jiaotong University , Xian , China
| | - Mingmei Wu
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey , New Jersey, NJ , USA
| | - Zhaoxia Duan
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey , New Jersey, NJ , USA
| | - Jing Lv
- Department of Biomedical Sciences, Florida State University College of Medicine , Tallahassee, FL , USA
| | - Wenjiao Tai
- Department of Biomedical Sciences, Florida State University College of Medicine , Tallahassee, FL , USA
| | - Hemamalini Renganathan
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey , New Jersey, NJ , USA
| | - Ruth Didier
- Department of Biomedical Sciences, Florida State University College of Medicine , Tallahassee, FL , USA
| | - Jinhua Li
- Department of Anatomy and Developmental Biology, Monash University , Clayton, VIC , Australia
| | - Dongming Sun
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey , New Jersey, NJ , USA
| | - Xiaoming Chen
- Institute of Translational Medicine, First Affiliated Hospital, Wenzhou Medical University , Wenzhou , China
| | - Xijing He
- Department of Orthopedic Surgery, The Second Hospital of Xian Jiaotong University , Xian , China
| | - Jianqing Fan
- Statistics Laboratory, Princeton University , Princeton, NJ , USA
| | - Wise Young
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey , New Jersey, NJ , USA
| | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine , Tallahassee, FL , USA
| |
Collapse
|
40
|
Forget MA, Voorhees JL, Cole SL, Dakhlallah D, Patterson IL, Gross AC, Moldovan L, Mo X, Evans R, Marsh CB, Eubank TD. Macrophage colony-stimulating factor augments Tie2-expressing monocyte differentiation, angiogenic function, and recruitment in a mouse model of breast cancer. PLoS One 2014; 9:e98623. [PMID: 24892425 PMCID: PMC4043882 DOI: 10.1371/journal.pone.0098623] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 05/06/2014] [Indexed: 02/07/2023] Open
Abstract
Reports demonstrate the role of M-CSF (CSF1) in tumor progression in mouse models as well as the prognostic value of macrophage numbers in breast cancer patients. Recently, a subset of CD14+ monocytes expressing the Tie2 receptor, once thought to be predominantly expressed on endothelial cells, has been characterized. We hypothesized that increased levels of CSF1 in breast tumors can regulate differentiation of Tie2- monocytes to a Tie2+ phenotype. We treated CD14+ human monocytes with CSF1 and found a significant increase in CD14+/Tie2+ positivity. To understand if CSF1-induced Tie2 expression on these cells improved their migratory ability, we pre-treated CD14+ monocytes with CSF1 and used Boyden chemotaxis chambers to observe enhanced response to angiopoietin-2 (ANG2), the chemotactic ligand for the Tie2 receptor. We found that CSF1 pre-treatment significantly augmented chemotaxis and that Tie2 receptor upregulation was responsible as siRNA targeting Tie2 receptor abrogated this effect. To understand any augmented angiogenic effect produced by treating these cells with CSF1, we cultured human umbilical vein endothelial cells (HUVECs) with conditioned supernatants from CSF1-pre-treated CD14+ monocytes for a tube formation assay. While supernatants from CSF1-pre-treated TEMs increased HUVEC branching, a neutralizing antibody against the CSF1R abrogated this activity, as did siRNA against the Tie2 receptor. To test our hypothesis in vivo, we treated PyMT tumor-bearing mice with CSF1 and observed an expansion in the TEM population relative to total F4/80+ cells, which resulted in increased angiogenesis. Investigation into the mechanism of Tie2 receptor upregulation on CD14+ monocytes by CSF1 revealed a synergistic contribution from the PI3 kinase and HIF pathways as the PI3 kinase inhibitor LY294002, as well as HIF-1α-deficient macrophages differentiated from the bone marrow of HIF-1αfl/fl/LysMcre mice, diminished CSF1-stimulated Tie2 receptor expression.
Collapse
Affiliation(s)
- Mary A. Forget
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Molecular Cellular and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Jeffrey L. Voorhees
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Sara L. Cole
- Campus Microscopy and Imaging Facility, The Ohio State University, Columbus, Ohio, United States of America
| | - Duaa Dakhlallah
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Ivory L. Patterson
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Amy C. Gross
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Leni Moldovan
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Xiaokui Mo
- The Center for Biostatistics, The Ohio State University, Columbus, Ohio, United States of America
| | - Randall Evans
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Clay B. Marsh
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Molecular Cellular and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Tim D. Eubank
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
41
|
Abstract
The CSF-1 receptor (CSF-1R) is activated by the homodimeric growth factors colony-stimulating factor-1 (CSF-1) and interleukin-34 (IL-34). It plays important roles in development and in innate immunity by regulating the development of most tissue macrophages and osteoclasts, of Langerhans cells of the skin, of Paneth cells of the small intestine, and of brain microglia. It also regulates the differentiation of neural progenitor cells and controls functions of oocytes and trophoblastic cells in the female reproductive tract. Owing to this broad tissue expression pattern, it plays a central role in neoplastic, inflammatory, and neurological diseases. In this review we summarize the evolution, structure, and regulation of expression of the CSF-1R gene. We discuss the structures of CSF-1, IL-34, and the CSF-1R and the mechanism of ligand binding to and activation of the receptor. We further describe the pathways regulating macrophage survival, proliferation, differentiation, and chemotaxis downstream from the CSF-1R.
Collapse
Affiliation(s)
- E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
42
|
PI3K p110γ deletion attenuates murine atherosclerosis by reducing macrophage proliferation but not polarization or apoptosis in lesions. PLoS One 2013; 8:e72674. [PMID: 23991137 PMCID: PMC3750002 DOI: 10.1371/journal.pone.0072674] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 07/17/2013] [Indexed: 02/02/2023] Open
Abstract
Atherosclerosis is an inflammatory disease regulated by infiltrating monocytes and T cells, among other cell types. Macrophage recruitment to atherosclerotic lesions is controlled by monocyte infiltration into plaques. Once in the lesion, macrophage proliferation in situ, apoptosis, and differentiation to an inflammatory (M1) or anti-inflammatory phenotype (M2) are involved in progression to advanced atherosclerotic lesions. We studied the role of phosphoinositol-3-kinase (PI3K) p110γ in the regulation of in situ apoptosis, macrophage proliferation and polarization towards M1 or M2 phenotypes in atherosclerotic lesions. We analyzed atherosclerosis development in LDLR−/−p110γ+/− and LDLR−/−p110γ−/− mice, and performed expression and functional assays in tissues and primary cells from these and from p110γ+/− and p110γ−/− mice. Lack of p110γ in LDLR−/− mice reduces the atherosclerosis burden. Atherosclerotic lesions in fat-fed LDLR−/−p110γ−/− mice were smaller than in LDLR−/−p110γ+/− controls, which coincided with decreased macrophage proliferation in LDLR−/−p110γ−/− mouse lesions. This proliferation defect was also observed in p110γ−/− bone marrow-derived macrophages (BMM) stimulated with macrophage colony-stimulating factor (M-CSF), and was associated with higher intracellular cyclic adenosine monophosphate (cAMP) levels. In contrast, T cell proliferation was unaffected in LDLR−/−p110γ−/− mice. Moreover, p110γ deficiency did not affect macrophage polarization towards the M1 or M2 phenotypes or apoptosis in atherosclerotic plaques, or polarization in cultured BMM. Our results suggest that higher cAMP levels and the ensuing inhibition of macrophage proliferation contribute to atheroprotection in LDLR−/− mice lacking p110γ. Nonetheless, p110γ deletion does not appear to be involved in apoptosis, in macrophage polarization or in T cell proliferation.
Collapse
|
43
|
Hamilton JA, Lacey DC, Turner A, de Kok B, Huynh J, Scholz GM. Hypoxia enhances the proliferative response of macrophages to CSF-1 and their pro-survival response to TNF. PLoS One 2012; 7:e45853. [PMID: 23029275 PMCID: PMC3446916 DOI: 10.1371/journal.pone.0045853] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Accepted: 08/23/2012] [Indexed: 01/18/2023] Open
Abstract
In chronic inflammatory lesions there are increased numbers of macrophages with a possible contribution of enhanced survival/proliferation due, for example, to cytokine action; such lesions are often hypoxic. Prior studies have found that culture in low oxygen can promote monocyte/macrophage survival. We show here, using pharmacologic inhibitors, that the hypoxia-induced pro-survival response of macrophages exhibits a dependence on PI3-kinase and mTOR activities but surprisingly is suppressed by Akt and p38 MAPK activities. It was also found that in hypoxia at CSF-1 concentrations, which under normoxic conditions are suboptimal for macrophage proliferation, macrophages can proliferate more strongly with no evidence for alteration in CSF-1 receptor degradation kinetics. TNF promoted macrophage survival in normoxic conditions with an additive effect in hypoxia. The enhanced hypoxia-dependent survival and/or proliferation of macrophages in the presence of CSF-1 or TNF may contribute to their elevated numbers at a site of chronic inflammation.
Collapse
Affiliation(s)
- John A Hamilton
- Arthritis and Inflammation Research Centre, University of Melbourne Department of Medicine, The Royal Melbourne Hospital Parkville, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
44
|
Wang Y, Piper MG, Marsh CB. The role of Src family kinases in mediating M-CSF receptor signaling and monocytic cell survival. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/abb.2012.35077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Jagger AL, Evans HG, Walter GJ, Gullick NJ, Menon B, Ballantine LE, Gracie A, Magerus-Chatinet A, Tiemessen MM, Geissmann F, Rieux-Laucat F, Taams LS. FAS/FAS-L dependent killing of activated human monocytes and macrophages by CD4+CD25- responder T cells, but not CD4+CD25+ regulatory T cells. J Autoimmun 2011; 38:29-38. [PMID: 22197557 DOI: 10.1016/j.jaut.2011.11.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 11/07/2011] [Accepted: 11/28/2011] [Indexed: 12/24/2022]
Abstract
Conclusive resolution of an immune response is critical for the prevention of autoimmunity and chronic inflammation. We report that following co-culture with autologous CD4+CD25- responder T cells, human CD14+ monocytes and monocyte-derived macrophages become activated but also significantly more prone to apoptosis than monocytes/macrophages cultured alone. In contrast, in the presence of CD4+CD25+ regulatory T cells (Tregs), monocytes and macrophages survive whilst adopting an anti-inflammatory phenotype. The induction of monocyte death requires responder T cell activation and cell-contact between responder T cells and monocytes. We demonstrate a critical role for FAS/FAS-L ligation in responder T cell-induced monocyte killing since responder T cells, but not Tregs, upregulate FAS-ligand (FAS-L) mRNA, and induce FAS expression on monocytes. Furthermore, responder T cell-induced monocyte apoptosis is blocked by neutralising FAS/FAS-L interaction, and is not observed when monocytes from an autoimmune lymphoproliferative syndrome (ALPS) patient with complete FAS-deficiency are used as target cells. Finally, we show that responder T cell-induced killing of monocytes is impaired in patients with active rheumatoid arthritis (RA). Our data suggest that resolution of inflammation in the course of a healthy immune response is aided by the unperturbed killing of monocytes with inflammatory potential by responder T cells and the induction of longer-lived, Treg-induced, anti-inflammatory monocytes.
Collapse
Affiliation(s)
- Ann L Jagger
- Centre for Molecular and Cellular Biology of Inflammation, Division of Immunology, Infection and Inflammatory Disease, King's College London, London SE1 1UL, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang Y, Mo X, Piper MG, Wang H, Parinandi NL, Guttridge D, Marsh CB. M-CSF induces monocyte survival by activating NF-κB p65 phosphorylation at Ser276 via protein kinase C. PLoS One 2011; 6:e28081. [PMID: 22216091 PMCID: PMC3245220 DOI: 10.1371/journal.pone.0028081] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 10/31/2011] [Indexed: 01/05/2023] Open
Abstract
Macrophage colony-stimulating factor (M-CSF) promotes mononuclear phagocyte survival and proliferation. The transcription factor Nuclear Factor-kappaB (NF-κB) is a key regulator of genes involved in M-CSF-induced mononuclear phagocyte survival and this study focused at identifying the mechanism of NF-κB transcriptional activation. Here, we demonstrate that M-CSF stimulated NF-κB transcriptional activity in human monocyte-derived macrophages (MDMs) and the murine macrophage cell line RAW 264.7. The general protein kinase C (PKC) inhibitor Ro-31-8220, the conventional PKCα/β inhibitor Gö-6976, overexpression of dominant negative PKCα constructs and PKCα siRNA reduced NF-κB activity in response to M-CSF. Interestingly, Ro-31-8220 reduced Ser276 phosphorylation of NF-κBp65 leading to decreased M-CSF-induced monocyte survival. In this report, we identify conventional PKCs, including PKCα as important upstream kinases for M-CSF-induced NF-κB transcriptional activation, NF-κB-regulated gene expression, NF-κB p65 Ser276 phosphorylation, and macrophage survival. Lastly, we find that NF-κB p65 Ser276 plays an important role in basal and M-CSF-stimulated NF-κB activation in human mononuclear phagocytes.
Collapse
Affiliation(s)
- Yijie Wang
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Xiaokui Mo
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Melissa G. Piper
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Hongmei Wang
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Narasimham L. Parinandi
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Denis Guttridge
- Department of Human Cancer Genetics, The Ohio State University, Columbus, Ohio, United States of America
| | - Clay B. Marsh
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
47
|
Dutra RC, Cola M, Leite DFP, Bento AF, Claudino RF, Nascimento AFZ, Leal PC, Calixto JB. Inhibitor of PI3Kγ ameliorates TNBS-induced colitis in mice by affecting the functional activity of CD4+CD25+FoxP3+ regulatory T cells. Br J Pharmacol 2011; 163:358-74. [PMID: 21244371 DOI: 10.1111/j.1476-5381.2011.01226.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Phosphoinositide 3-kinase-γ (PI3Kγ) is implicated in many pathophysiological conditions, and recent evidence has suggested its involvement in colitis. In the present study, we investigated the effects of AS605240, a relatively selective PI3Kγ inhibitor, in experimental colitis and its underlying mechanisms. EXPERIMENTAL APPROACH Acute colitis was induced in mice by treatment with trinitrobenzene sulphonic acid (TNBS), and the effect of AS605240 on colonic injury was assessed. Pro-inflammatory mediators and cytokines were measured by immunohistochemistry, elisa, real time-polymerase chain reaction and flow cytometry. KEY RESULTS Oral administration of AS605240 significantly attenuated TNBS-induced acute colitis and diminished the expression of matrix metalloproteinase-9 and vascular endothelial growth factor. The colonic levels and expression of IL-1β, CXCL-1/KC, MIP-2 and TNF-α were also reduced following therapeutic treatment with AS605240. Moreover, AS605240 reduced MIP-2 levels in a culture of neutrophils stimulated with lipopolysaccharide. The mechanisms underlying these actions of AS605240 are related to nuclear factor-κ (NF-κB) inhibition. Importantly, the PI3Kγ inhibitor also up-regulated IL-10, CD25 and FoxP3 expression. In addition, a significant increase in CD25 and FoxP3 expression was found in isolated lamina propria CD4+ T cells of AS605240-treated mice. The effect of AS605240 on Treg induction was further confirmed by showing that concomitant in vivo blockade of IL-10R significantly attenuated its therapeutic activity. CONCLUSIONS AND IMPLICATIONS These results suggest that AS605240 protects mice against TNBS-induced colitis by inhibiting multiple inflammatory components through the NF-κB pathway while simultaneously inducing an increase in the functional activity of CD4+CD25+ Treg. Thus, AS605240 may offer a promising new therapeutic strategy for the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- R C Dutra
- Department of Pharmacology, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Kogan M, Haine V, Ke Y, Wigdahl B, Fischer-Smith T, Rappaport J. Macrophage colony stimulating factor regulation by nuclear factor kappa B: a relevant pathway in human immunodeficiency virus type 1 infected macrophages. DNA Cell Biol 2011; 31:280-9. [PMID: 21895511 DOI: 10.1089/dna.2011.1357] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Macrophage colony stimulating factor (M-CSF) is a cytokine that promotes monocyte differentiation and survival. When overexpressed, M-CSF contributes to pathology in a wide variety of diseases, including osteoporosis, obesity, certain human cancers, and in human immunodeficiency virus type 1 (HIV-1) infection, particularly with respect to monocyte/macrophage infection and the development of HIV-1 associated central nervous system disorders. In this study, our aim was to expand the current knowledge of M-CSF regulation, focusing on nuclear factor kappa B (NF-κB), a transcription factor playing a prominent role during inflammation and HIV-1 infection. Our results suggest that tumor necrosis factor alpha (TNF-α) promotes M-CSF secretion in primary macrophages and activates the -1310/+48 bp M-CSF promoter in Mono-Mac 1 cells. Inhibitors of the NF-κB pathway diminish this response. We identified four putative NF-κB and four CCAAT-enhancer-binding protein beta binding sites within the M-CSF promoter. Our findings, using promoter constructs mutated at individual NF-κB sites within the M-CSF promoter region, suggest that these sites are redundant with respect to NF-κB regulation. TNF-α treatment promoted NF-κB p65 binding to the M-CSF promoter in phorbol 12-myristate 13-acetate (PMA) treated U937 cells chronically infected with HIV-1 (U1 cells), but not in PMA treated uninfected U937 cells, suggesting that the presence of HIV-1 increases the NF-κB response. In conclusion, our findings demonstrate that NF-κB induces M-CSF expression on a promoter level via multiple functional NF-κB binding sites and that this pathway is likely relevant in HIV-1 infection of macrophages.
Collapse
Affiliation(s)
- Michael Kogan
- Department of Neuroscience, Center for Neurovirology, School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | |
Collapse
|
49
|
Braun T, Zwerina J. Positive regulators of osteoclastogenesis and bone resorption in rheumatoid arthritis. Arthritis Res Ther 2011; 13:235. [PMID: 21861862 PMCID: PMC3239343 DOI: 10.1186/ar3380] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bone destruction is a frequent and clinically serious event in patients with rheumatoid arthritis (RA). Local joint destruction can cause joint instability and often necessitates reconstructive or replacement surgery. Moreover, inflammation-induced systemic bone loss is associated with an increased fracture risk. Bone resorption is a well-controlled process that is dependent on the differentiation of monocytes to bone-resorbing osteoclasts. Infiltrating as well as resident synovial cells, such as T cells, monocytes and synovial fibroblasts, have been identified as sources of osteoclast differentiation signals in RA patients. Pro-inflammatory cytokines are amongst the most important mechanisms driving this process. In particular, macrophage colony-stimulating factor, RANKL, TNF, IL-1 and IL-17 may play dominant roles in the pathogenesis of arthritis-associated bone loss. These cytokines activate different intracellular pathways to initiate osteoclast differentiation. Thus, over the past years several promising targets for the treatment of arthritic bone destruction have been defined.
Collapse
Affiliation(s)
- Tobias Braun
- Department of Medicine 3, University of Erlangen-Nuremberg, Erlangen, 91054 Germany
| | | |
Collapse
|
50
|
Pedrera M, Gómez-Villamandos JC, Risalde MA, Molina V, Sánchez-Cordón PJ. Characterization of apoptosis pathways (intrinsic and extrinsic) in lymphoid tissues of calves inoculated with non-cytopathic bovine viral diarrhoea virus genotype-1. J Comp Pathol 2011; 146:30-9. [PMID: 21612789 DOI: 10.1016/j.jcpa.2011.03.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 03/24/2011] [Accepted: 03/29/2011] [Indexed: 11/26/2022]
Abstract
Previous studies have shown that activation of effector caspase-3 is associated with the apoptosis of lymphocytes occurring during infection with bovine viral diarrhoea virus (BVDV); however, the regulation of the apoptosis pathways that induce cell death via activation of effector caspase-3 has not yet been clarified. The aim of this study was to examine immunohistochemically the expression of cleaved caspase (CCasp)-8 (initiator caspase of the extrinsic pathway), CCasp9 (initiator caspase of the intrinsic pathway) and Bcl-2 (an anti-apoptotic marker) in gut-associated lymphoid tissue (GALT) of the ileum from calves inoculated with a non-cytopathic strain of BVDV genotype-1. CCasp8 had similar expression to that of CCasp3. In interfollicular T-cell areas there was moderate apoptosis and evidence of moderate activation of initiator caspase-8. In B-cell follicles there was marked lymphocyte apoptosis and evidence of intense caspase-8 activation, highlighting the potentially major role of the extrinsic pathway in lymphocyte apoptosis in the GALT during BVDV infection. Additionally, there was a significant decrease in the number of CCasp9(+) cells from the start of the experiment and this was linked to inactivation of caspase-9. Therefore, the intrinsic pathway may play only a minor role in the induction of lymphocyte apoptosis. Finally, the observed overexpression of Bcl-2 protein could play a major role in protecting lymphocytes in the T-cell areas against apoptosis, while low levels of Bcl-2 expression could be associated with the follicular lymphocyte apoptosis occurring during BVDV infection.
Collapse
Affiliation(s)
- M Pedrera
- Department of Comparative Pathology, Veterinary Faculty, University of Córdoba, Edificio Sanidad Animal, Campus de Rabanales, 14014 Córdoba, Spain
| | | | | | | | | |
Collapse
|